Author’s Accepted Manuscript Metabolic engineering in the host Yarrowia lipolytica Ahmad M. Abdel-Mawgoud, Kelly A. Markham, Claire M. Palmer, Nian Liu, Gregory Stephanopoulos, Hal S. Alper www.elsevier.com/locate/ymben
PII: DOI: Reference:
S1096-7176(18)30273-8 https://doi.org/10.1016/j.ymben.2018.07.016 YMBEN1445
To appear in: Metabolic Engineering Received date: 3 July 2018 Revised date: 23 July 2018 Accepted date: 24 July 2018 Cite this article as: Ahmad M. Abdel-Mawgoud, Kelly A. Markham, Claire M. Palmer, Nian Liu, Gregory Stephanopoulos and Hal S. Alper, Metabolic engineering in the host Yarrowia lipolytica , Metabolic Engineering, https://doi.org/10.1016/j.ymben.2018.07.016 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting galley proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Metabolic engineering in the host Yarrowia lipolytica Ahmad M. Abdel-Mawgouda,1, Kelly A. Markhamb,1, Claire M. Palmerc,1, Nian Liua,1, Gregory Stephanopoulosa,*, Hal S. Alperb,c,* a
Department of Chemical Engineering Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge, Massachusetts 02139
b
McKetta Department of Chemical Engineering The University of Texas at Austin 200 E Dean Keeton St. Stop C0400 Austin, Texas 78712
c
Institute for Cellular and Molecular Biology The University of Texas at Austin 2500 Speedway Avenue Austin, Texas 78712 E-mail:
[email protected] E-mail:
[email protected] *
*
Co-Corresponding author: Phone: (617) 253-4583,Mailing Address: Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139
Co-Corresponding author: Phone: (512) 471-4417, Fax: (512) 471-7060, Mailing Address: 200 E Dean Keeton St. Stop C0400 Austin, TX 78712
Abstract The nonconventional, oleaginous yeast, Yarrowia lipolytica is rapidly emerging as a valuable host for the production of a variety of both lipid and nonlipid chemical products. While the unique genetics of this organism pose some challenges, many new metabolic engineering tools have emerged to facilitate improved genetic manipulation in this host. This review establishes a case for Y. lipolytica as a premier metabolic engineering host based on innate metabolic capacity, emerging synthetic tools, and engineering examples. The metabolism underlying the lipid accumulation phenotype of this yeast as well as high flux through acyl-CoA
1
Authors contributed equally to this work
1
precursors and the TCA cycle provide a favorable metabolic environment for expression of relevant heterologous pathways. These properties allow Y. lipolytica to be successfully engineered for the production of both native and nonnative lipid, organic acid, sugar and acetylCoA derived products. Finally, this host has unique metabolic pathways enabling growth on a wide range of carbon sources, including waste products. The expansion of carbon sources, together with the improvement of tools as highlighted here, have allowed this nonconventional organism to act as a cellular factory for valuable chemicals and fuels. Keywords: Yarrowia lipolytica, metabolic engineering, synthetic biology, oleochemicals, biofuel 1. Introduction Yarrowia lipolytica is emerging as the model non-conventional oleaginous yeast. As an organism with “generally regarded as safe” (GRAS) status (Groenewald et al., 2014), it has been widely recognized as a potential industrial workhorse for the production of lipid-based biofuels and oleochemicals (Markham et al., 2017). In particular, Y. lipolytica is well suited for industrial production of oleochemicals as wildtype strains can accumulate lipids up to 70% of dry biomass (Beopoulos and Nicaud, 2012). Key metabolic traits that contribute to this oleaginous phenotype include high acetyl-CoA flux, high tricarboxylic acid (TCA) cycle flux, and lack of fermentative capacity (Christen and Sauer, 2011; Kavšček et al., 2015). Moreover, Y. lipolytica has the ability to utilize diverse protein and hydrophobic substrates, which may be provided as cheap renewable carbon sources, and it grows at a wide range of pH and salinity conditions (Bankar et al., 2009; Michely et al., 2013). With these unique metabolic traits and recently developed metabolic engineering tools, this industrial host shows great promise for economic and renewable production of a plethora of
2
new products in the future. As such, this article reviews Y. lipolytica research, discussing the current understandings of its biology as well as achievements in the metabolic engineering of this organism. We first introduce basic microbiological features of Y. lipolytica and their relevance to metabolic engineering. Next, we discuss the genetic engineering and systems biology tools that are available for this organism to date. Following this, we discuss past successes that demonstrate the full potential of Y. lipolytica as a host for microbial production of lipid derived chemicals, organic acids, sugar alcohols, and acetyl-CoA derived products. Additionally, we highlight improvements in the metabolic engineering for diverse carbon utilization. Finally, we conclude with perspectives on the future of engineering in Y. lipolytica.
2. Y. lipolytica background and basics As natural niches for Y. lipolytica, this organism is readily found in lipid and/or protein rich substrates, such as cheeses (e.g. Camembert and blue-veined cheeses), dairy products (Roostita and Fleet, 1996), as well as meat and sausages (Fickers et al., 2005; Groenewald et al., 2014; Jacques and Casaregola, 2008). This organism is found to live in soil, sewage, and oilpolluted environments (Hassanshahian et al., 2012). These chemical habitats align well with an efficient metabolic capacity to hydrolyze lipids via secreted lipases and esterases, assimilate hydrocarbons and fatty acids via terminal, β-, and ω-oxidation pathways (Abghari and Chen, 2014; Thevenieau et al., 2010), as well as express and secrete extracellular proteases (Fabre et al., 1991). This efficient protein degradation and hydrophobic substrate catabolism seen in Y. lipolytica is supported by a host of genes encoding essential functions (Gaillardin et al., 2013). For example, Y. lipolytica has 16 acylglycerol lipases, 12 cytochrome P450s for hydrocarbon and fatty acid oxidation, 14 fatty acid transporters, as well as 38 aspartyl proteases and 15 serine
3
proteases (Gaillardin et al., 2013). As a result, these traits make this yeast an especially interesting organism to study and engineer.
2.1 Taxonomy and morphology Y. lipolytica, formerly classified as Candida lipolytica or Saccharomycopsis lipolytica (Esser and Stahl, 1976), is a yeast belonging to the Ascomycota phylum, Dipodascaceae family, Fungi kingdom (Kurtzman, 2011). It is denoted as a “non-conventional yeast” with respect to its distinctive genome structure and its large phylogenetic distance relative to other yeasts while sharing common properties with molds and higher eukaryotes (Barth and Gaillardin, 1996; Barth and Gaillardin, 1997; Dujon et al., 2004; Torres-Guzmán and Domínguez, 1997). Y. lipolytica is a heterothallic yeast commonly found in nature in either of the two haploid mating types, MatA or MatB, although diploid strains were occasionally isolated (Knutsen et al., 2007). With regard to morphology, dimorphism is one of the characteristic features of Y. lipolytica. Specifically, this organism can exist in either the yeast form or the pseudo-/septate hyphae (non-branching) forms (Barth and Gaillardin, 1996; Kawasse et al., 2003). Yeast-tohyphae ratio partly contributes to the diverse colony morphologies of Y. lipolytica and is affected by medium and growth conditions as well as by genetic factors (Barth and Gaillardin, 1996). As a result, regulation of yeast-to-hyphae transition is suggested to be complex and multifactorial and induced as a survival mechanism in response to stressful environmental and nutritional conditions (Bellou et al., 2014; Gancedo, 2001; Hurtado et al., 2000; Morales-Vargas et al., 2012; O'Shea and Walsh, 2000; Pérez-Campo and Domínguez, 2001; Ruiz-Herrera and Sentandreu, 2002; Torres-Guzmán and Domínguez, 1997). The morphological state of Y. lipolytica has profound impacts on bioreactor performance. Since yeast forms are associated with
4
better rheological properties, they are preferred to hyphal forms in submerged fermentation (Timoumi et al., 2017). On the other hand, hyphae forms may be desired in solid-state fermentations (Bellou et al., 2014). In a laboratory setting, colonies of Y. lipolytica are of creamy texture and show a convoluted pale white matte surface (Figure 1), whereas traditional yeast form rich colonies appear smooth and glistening (Heslot, 1990). Under a normal light microscope, cells of Y. lipolytica appear spherical, ellipsoidal, or elongated with typical dimensions of 3-5 X 3.3-15 µm and are arranged singly, in pairs, or clustered in groups (Kurtzman, 2011) (Figure 2). One particularly characteristic feature of Y. lipolytica is its possession of large intracellular lipid bodies. Lipid bodies, also called oleosomes, are neutral-lipid storage organelles (0.65 to 2.5 µm) (Athenstaedt, 2010) that are easily discernable using fluorescence microscopy (Figure 2) (Athenstaedt, 2010). In this organism, the lipid bodies are comprised mainly of triacylglycerides (TAG) as the inner core and, to a lesser extent, steryl esters (SE) as the outer core (Athenstaedt et al., 2006). The entire structure is encapsulated by a protein-embedded phospholipid monolayer (Beopoulos et al., 2009; Czabany et al., 2008). In general, lipid body formation is induced under unfavorable nutritional conditions, including nitrogen limitation or limited oxygen supply (Kavšček et al., 2015). These conditions direct metabolism towards energy storage as TAGs, which is part of a complex system of fatty acid homeostasis (Garay et al., 2014). Moreover, lipid body formation is suggested to play a role in free fatty acid detoxification by segregating these compounds away from the general cytosol and into a membrane-bound compartment (Athenstaedt, 2010). Overall, lipid bodies serve as dynamic storage organelles since the yeast can remobilize fatty acids out of lipid bodies by TAG lipases
5
and steryl ester hydrolases in response to its needs for biomass precursors and energy supply (Abghari and Chen, 2014).
2.2 Growth conditions Y. lipolytica is a strictly aerobic, non-fermentative yeast (Barth and Gaillardin, 1996) and thus requires good aeration for optimal growth, a potential limitation in some industrial fermentation settings. The recommended temperature for cultivation is 25-30 °C, but growth can occur at refrigeration temperature (Fickers et al., 2005; Kurtzman, 2011) and rarely even up to 34 °C (Barth and Gaillardin, 1996). Its inability to grow at temperatures above 34 °C and limited capacity to cause only mild, self-limiting infections in severely immunocompromised patients are amongst the reasons behind its GRAS status (Groenewald et al., 2014; Jacques and Casaregola, 2008). Y. lipolytica is exceedingly tolerant to a wide range of pH values, including highly acidic environments (Egermeier et al., 2017). However, it is interesting that across this pH range, the metabolic (Egermeier et al., 2017) and morphologic (Timoumi et al., 2017) profiles differ significantly. In terms of carbon sources, Y. lipolytica has a characteristic ability to assimilate a wide range of unusual hydrophobic and hydrophilic carbon sources. Hydrophilic carbon sources include two monosaccharides, namely glucose and fructose (Michely et al., 2013); the amino sugar N-acetyl-D-glucosamine; some alcohols, such as ethanol (up to 3%) but not methanol; many polyols, e.g. glycerol, erythritol, mannitol, glucitol; many organic acids, e.g. acetate (up to 0.4%), lactate, succinate and citrate(Barth and Gaillardin, 1997). The hydrophobic substrates that can be utilized by Y. lipolytica are alkanes (C12 to C16 but not C8 and C10), alkenes (Barth and Gaillardin, 1996; Kurtzman, 2011), fatty acids, fatty acid methyl esters, and triglycerides
6
(Michely et al., 2013). Amongst these carbon sources is the ability to utilize crude glycerol, an extremely low cost ($ 0.05 per pound) byproduct of the oil and biodiesel industries (Yang et al., 2012). Additional waste products including used frying oils have been explored as alternative, cheap carbon sources (Domínguez et al., 2010). Further engineering efforts (described later) are expanding the array of carbon sources. As for nitrogen sources, Y. lipolytica assimilates inorganic ammonium (but not nitrates), usually in the chloride or sulfate salt forms (Fickers et al., 2004; Kurtzman, 2011). More options are available for Y. lipolytica with regard to organic nitrogen sources including both defined ones, such as urea, and complex ones, such as casamino acids, peptones, tryptones, and yeast extract (Fickers et al., 2004). Interestingly, human urine, which contains urea at 1-2% w/v, has been showed to be an effective and cheap nitrogen source for Y. lipolytica (Brabender et al., 2018). In general, the nature and concentration of nitrogen source play a significant role in growth and lipid production in Y. lipolytica as discussed later. Vitamin requirements are also an important consideration in the cultivation of strains. Y. lipolytica is auxotrophic to thiamine (vitamin B1) only, unlike other members of the Yarrowia clade which are auxotrophic to multiple vitamins (Barth and Gaillardin, 1996; Kurtzman, 2011). More simply, Y. lipolytica can grow in minimal medium without YNB added, but requires supplementation of thiamine (at 0.4-1 mg/L) together with some trace elements (Egermeier et al., 2017; Jost et al., 2015). No studies have systemically investigated, however, the complete trace element requirements of Y. lipolytica (Gasmi et al., 2011). A good starting point for such a study could be inspired, however, from the composition of trace elements provided in the commercial trace elements solution 2 (Celińska and Grajek, 2013) or YNB in order to develop a standard, less expensive defined minimal medium.
7
3. Genetics The metabolic engineering of any microorganism often requires genetic manipulations, which cannot be done without a detailed understanding of its genome structure as well as its gene expression machinery. Y. lipolytica is no exception, and indeed, the community has accumulated considerable knowledge about its genetics. This section is dedicated to the review of relevant genetic information in Y. lipolytica.
3.1 Genomic elements The genome of Y. lipolytica is composed of 6 nuclear chromosomes (A to F) (Dujon et al., 2004; Pomraning and Baker, 2015) and 1 mitochondrial chromosome (Kerscher et al., 2001). Both haploid and diploid strains of Y. lipolytica propagate stably in laboratory settings (Knutsen et al., 2007), with haploid strains more favored in lab settings. The genomes of the three widely studied strains of Y. lipolytica, CLIB122 (Dujon et al., 2004), W29 (CLIB89) (Pomraning and Baker, 2015), and PO1f (Liu and Alper, 2014) together with certain derivative strains have been sequenced and made available at GeneBank of the NCBI website (https://www.ncbi.nlm.nih.gov/genome/genomes/194). The genome sequence of Y. lipolytica CLIB122 is also found through the website of Genome Resources for Yeast Chromosomes (http://gryc.inra.fr/index.php?page=home). The total genome size of Y. lipolytica strains CLIB122 or W29, including the mitochondrial chromosome, is 20.5 Mb with a net G-C content of 49%, which are 1.7 and 1.28 times larger than those of S. cerevisiae, respectively (Dujon et al., 2004).
8
Genome annotations of CLIB122 and W29 show a total of 7144 and 8746 genes, respectively. Of these genes, 6472 and 7949 are protein-coding genes (open reading frames, ORFs) in CLIB122 and W29, respectively. Such numbers of ORFs are unexpectedly low given the remarkably larger genome size of Y. lipolytica (20.5 Mb) compared to that of S. cerevisiae (12 Mb) (Dujon et al., 2004). This obviously reflects on a lower gene density—a value that is 46.3% in Y. lipolytica (i.e. 1 gene/3 Kb) compared to 70.3% (1 gene/2 Kb) in S. cerevisiae (Dujon et al., 2004). These large intergenic regions have allowed for identification of 11 chromosomal sites suitable for gene integration in intergenic regions surrounded by highly expressed neighboring genes, according to a previous transcriptomic study (Morin et al., 2011), while distant enough from these native neighboring genes not to cause any possible perturbation to their function (Holkenbrink et al., 2018). A particularly important component of the chromosomal DNA for metabolic engineering in Y. lipolytica is the ribosomal DNA (rDNA), as it is often targeted for homology-based multicopy gene integration. Y. lipolytica has 7 subtelomeric loci of dispersed rDNA repeats, which is distinctive than most hemiascomycetes where there are only 1 to 3 intrachromosomal clustered rather than dispersed rDNA repeat (Dujon et al., 2004). At least 105 copies of 5S rDNA are dispersed throughout the genome of Y. lipolytica (Dujon et al., 2004). Previously, there was an attempt to use these rDNA repeats (8.7 Kb) to construct multicopy gene integration vectors (Juretzek et al., 2001; Le Dall et al., 1994). However, the method proved ineffective due to concerns regarding the number and stability of gene copies in the genome (Le Dall et al., 1994). Nonetheless, this approach has been used effectively to engineer these cells for improved production of products (Blazeck et al., 2013a; Celińska and Grajek, 2013; Liu et al., 2017a).
9
As mentioned before, one chromosome of Y. lipolytica resides in the mitochondrion. The mitochondrial DNA (mtDNA) of Y. lipolytica is 47.9 Kb long and codes for the 14 hydrophobic subunits of respiratory chain complexes, the large and small rRNAs, and 27 tRNAs (Kerscher et al., 2001). Being a strictly aerobic non-fermentative yeast, Y. lipolytica shows the most powerful translational efficiency of mitochondrial genes related to aerobic respiration (Man and Pilpel, 2007) and a strong TCA cycle flux (Christen and Sauer, 2011). Accordingly, Y. lipolytica cannot survive mitochondrial mutations affecting respiratory chains and hence this yeast is classified as a petite mutant-negative species (Bakkaiova et al., 2014; Savická and Šilhánková, 1995). The high-expression genetic environment of mtDNA provide an optimum place for integration of heterologous genes that are required to be expressed in this organelle. Nonetheless, genetic manipulations of mtDNA is still challenging (Gammage et al., 2017). Recently, several successful trials in Y. lipolytica (Isakova et al., 2015) have provided promise for more efficient mitochondrial genome engineering in Y. lipolytica.
3.2 Translational machinery Regarding its translational machinery, Y. lipolytica possesses 510 genes coding for tRNA, which is the largest number amongst yeasts and 1.86 times higher than that of S. cerevisiae (Dujon et al., 2004). In Y. lipolytica, the tRNA genes code for the same 44-tDNA set as higher eukaryotes. Uniquely, among all eukaryotic systems, 26 of these tRNA contain introns in Y. lipolytica (Dujon et al., 2004; Neuvéglise et al., 2013). More interestingly, Y. lipolytica harbors ~50 copies of hybrid dicistronic co-transcribed tRNA-5S rRNA genes (Neuvéglise et al., 2013) in addition to the solo tRNA and tandem-tRNA (di-cistronic) conventionally found in hemiascomycetes (Neuvéglise et al., 2005). This implies that Y. lipolytica possesses an efficient
10
and unique tRNA processing/maturation machinery which was recently exploited in engineering tRNA-gRNA fusion for CRISPR/Cas9-mediated genome editing in Y. lipolytica (Schwartz et al., 2016).
3.3 mRNA processing Hemiascomycetes are generally intron-deficient compared to higher eukaryotes (Stajich et al., 2007). Nonetheless, Y. lipolytica has the most intron-rich genome in yeasts with at least 16% of its genes containing introns (Mekouar et al., 2010). Interestingly, the length and number of these introns per gene vary remarkably across the genome. As such, the average spliceosomal intron length in Y. lipolytica is ~280 bp (Stajich et al., 2007). Yet, there are 16 introns with 1 to 3.4 Kb whereas 300 introns are less than 100 bp (Gaillardin et al., 2013). As for the number of introns per single gene, 87 genes contain multiple introns in Y. lipolytica which is more than 20 times higher than that in S. cerevisiae (where only 4 genes have multiple introns) (Thevenieau et al., 2009). In Y. lipolytica, mRNA splicing proceeds via both conventional (Gaillardin et al., 2013; Stajich et al., 2007) and alternative splicing (Gaillardin et al., 2013; Neuvéglise et al., 2005). Interestingly, intron-containing genes can show higher expression profiles than intronless versions of the same genes (Le Hir et al., 2003) as demonstrated recently (Hong et al., 2012; Tai and Stephanopoulos, 2013). Specifically, one study showed a 5-fold increase in the expression of β-glucuronidase gene under the fructose-bisphosphate aldolase-1 promoter (pFBA1) compared to the intronless version of the same gene (Hong et al., 2012). Another study reported a 17-fold increase in expression of the intron-containing β-galactosidase gene under translation elongation factor-1α promoter (pTEF1in) compared to the intronless version of the same gene (Tai and
11
Stephanopoulos, 2013). The abundance of introns present in Y. lipolytica leads to interesting queries to improve our future metabolic engineering efforts in this host.
3.4 DNA repair and recombination Similar to other eukaryotes (Ma et al., 2003), mechanisms involved in DNA doublestrand break (DSB) repair in Y. lipolytica include homology-directed recombination (HR) (Richard et al., 2005) and non-homologous end joining (NHEJ) (Daley et al., 2005; Richard et al., 2005). Although Y. lipolytica is capable of both modes of DNA repair, NHEJ is the dominant form of DNA repair (Verbeke et al., 2013) meaning that donor DNA is randomly integrated into the genome. From a genetic engineering point of view, a more specific loci targeting is preferred to decrease the heterogeneity that occurs with random integration (Madzak and Beckerich, 2013). NHEJ is enabled by the heterodimer protein complex Ku70/Ku80 (Lustig, 1999). Knockout of the genes encoding this complex resulted in an increased rate of selected cells that had undergone HR in Y. lipolytica (Kretzschmar et al., 2013; Verbeke et al., 2013; Bredeweg et al., 2017). Recently, CRISPR-Cas9 mediated gene integration technology has been adapted to Y. lipolytica (Gao et al., 2016b; Schwartz et al., 2016) and was associated with HR rate of about 50 to 70% (Schwartz et al., 2017b). These methods facilitate the isolation of cells with locusspecific modifications, improving precision of genetic engineering in Y. lipolytica.
4. Metabolic engineering and synthetic biology tools Developing efficient and predictable synthetic biology tools is essential to advancing the scope of metabolic engineering possibilities in any organism. In this section, we discuss the genetic tools that have been developed that help facilitate metabolic engineering in Y. lipolytica. 12
We then describe control elements that allow for modulating gene expression. Following control strategies, we discuss advances in emerging tools like CRISPR-Cas9. Finally, we demonstrate the development of effective models that will guide future strain design efforts. Collectively, these tools will enable metabolic engineering efforts catapulting Y. lipolytica chemical production to industrial successes.
4.1 Basic genetic tools As a non-conventional yeast, enabling metabolic engineering of Y. lipolytica has required the non-trivial development of basic genetic tools. Due to its propensity for NHEJ, traditional methods for gene expression on episomal plasmids are limited, but we discuss the vectors that have been developed for this purpose. We investigate optimized transformation methods allowing for the integration of exogenous DNA. Finally, we report on the selectable markers that have been effectively used in Y. lipolytica.
4.1.1 Episomal plasmids have been developed, but are generally unstable As a first step in making Y. lipolytica genetically tractable, episomal plasmids were investigated. Episomal plasmids are convenient tools for rapid genetics, but Y. lipolytica is devoid of any identifiable native plasmid (Heslot, 1990). Not surprisingly, Y. lipolytica thus still struggles to maintain any artificially transformed replicative plasmids (especially at high copy) despite efforts from several groups. An early patent describes an autonomously replicating sequence (ARS) that is effective in Y. lipolytica although unstable in mitosis (Fournier et al., 1993b). Further efforts demonstrated that two previously discovered ARS sequences (Fournier et al., 1993a; Fournier et al., 1991) were actually comprised of two essential elements--an origin of
13
replication and a centromere (Vernis et al., 1997; Vernis et al., 2001). These ARS sequences are quite large compared to other hosts and thus more minimal regions have been explored (Yamane et al., 2008a; Yamane et al., 2008b). Recent work has improved copy number by 80% through fusing promoters upstream of the centromeric region to enable a dynamic expression range of 2.7-fold (Liu et al., 2014). Overall, stable episomal plasmid expression is still strongly limiting in this host.
4.1.2 Transformation Given the limitations of episomal plasmid expression there has been a focus on improving protocols for integrative transformation. While these methods can be used to transform episomal plasmids, the transformation efficiencies reported here refer specifically to integrative transformations. Y. lipolytica uptakes exogenous DNA with moderate efficiency compared to S. cerevisiae, which can regularly achieve transformation efficiencies exceeding 107 transformants per µg DNA (Kawai et al., 2010). Most transformation methods in Y. lipolytica use a chemical transformation technique starting with a lithium acetate treatment, followed by incubation in polyethylene glycol (PEG), dithiothreitol (DTT), and DNA at an elevated temperature. An early protocol uses this lithium acetate treatment, but requires an excessive amount of sonicated carrier DNA to achieve 104 transformants per µg DNA per 108 viable cells (Davidow et al., 1985). Without carrier DNA, this protocol results in less than 20 transformants (Davidow et al., 1985). Later studies have optimized this method by developing a one-step protocol where cells are directly prepared from a YPD plate (Chen et al., 1997), increasing site specific integration by using a strain with a zeta docking platform (Bordes et al., 2007), and adapting this protocol for high-throughput transformation in a 96-well plate (Leplat et al., 2015). 14
Each of these methods reports maximum transformation efficiencies generally ranging from 103 – 104 transformants per µg DNA, but suffer from limited efficiency for site-directed integration due to Y. lipolytica’s natural propensity for NHEJ. In addition to chemical transformation methods, an electroporation method for Y. lipolytica transformation has been developed with efficiencies up to 104 transformants per µg DNA (Wang et al., 2011). Collectively, these results demonstrate the genetic tractability of Y. lipolytica, though there is room to improve these transformation efficiencies, specifically for site-directed integrations.
4.1.3 A plethora of markers allow for incorporation of exogenous DNA in Y. lipolytica To offset the lack of strong episomal elements, a variety of different selective markers have been developed for use in Y. lipolytica genome engineering including both auxotrophic and dominant selection schemes. Straightforward auxotrophic selection is well documented in Y. lipolytica (Barth and Gaillardin, 1996; Madzak et al., 2004). In particular, the commonly used PO1 series of Y. lipolytica strains provide useful auxotrophies (Madzak et al., 2000). Additionally, a defective URA3 has been developed that allows for multi-copy integrations using a single transformation, up to 60 copies (Le Dall et al., 1994). Although Y. lipolytica is natively resistant to many commonly used antibiotics (Gaillardin and Ribet, 1987), dominant selection schemes have been reported for resistance to phleomycin, hygromycin, nourseothricin, chlorimuron ethyl, and mycophenolic acid (Wagner et al., 2018b). An alternative dominant marker has been developed that allows for selection with sucrose through expression of the SUC2 gene from S. cerevisiae (Nicaud et al., 1989). Together, this abundance of selection schemes allow for incorporation of exogenous DNA into Y. lipolytica
15
enabling complex pathway engineering. Moreover, marker rescue systems like Cre-lox recombination have been implemented in this host (Fickers et al., 2003). Likewise, a collection of vector sets and associated DNA assembly tools have been described and reviewed in recent years (Holkenbrink et al., 2018; Celińska et al., 2017; Gao et al., 2014; Markham and Alper, 2018; Wong et al., 2017). Collectively, these basic genetic tools lay the foundation for all metabolic engineering in Y. lipolytica.
4.2 Control elements of gene expression After enabling metabolic engineering with the basic genetic tools reported above, modulation of expression is achievable through control elements like promoters and terminators. At the most basic level, native promoters have different strengths leading to a variety of different expression levels. A common strategy for increasing promoter strength, particularly for Y. lipolytica, is to identify and concatenate upstream activating sequences in front of a core promoter (Blazeck et al., 2011; Blazeck et al., 2013b; Madzak et al., 1999; Madzak et al., 2000). Adding an additional level of control is achievable by hybridizing inducible elements such as for oleic acid (Shabbir Hussain et al., 2016; Shabbir Hussain et al., 2017), n-decane (Sumita et al., 2002), and erythritol (Trassaert et al., 2017). An added level of control is achievable by using synthetic terminators (Curran et al., 2015). For further information, control elements for Y. lipolytica have been recently reviewed in greater detail (Markham and Alper). Collectively, these control elements allow for modulating gene expression to enable more complex metabolic engineering strategies.
4.3 Emerging tools for rapid engineering
16
A series of emerging tools have demonstrated the potential of synthetic biology to revolutionize metabolic engineering efforts. First, we discuss how CRISPR-Cas9 has changed the scope of engineering possibilities in Y. lipolytica. Then, we investigate the utility of transposase systems for enabling multi-copy integrations and functional genomics. Together, these systems demonstrate the ever-evolving future of metabolic engineering in Y. lipolytica.
4.3.1 CRISPR-Cas9 is a powerful tool for metabolic engineering in Y. lipolytica Recent advances in CRISPR-Cas9 based genome engineering are beginning to improve prospects for engineering Y. lipolytica. Specifically, CRISPR-Cas9 was first implemented by Schwartz et al. through the use of an RNA polymerase II promoter or synthetic RNA polymerase III promoter with hammerhead and HDV ribozymes to drive sgRNA expression enabling single gene disruptions of pex10, mfe1, and ku70 with high efficiency (Schwartz et al., 2016). This work was later expanded to demonstrate multiplex editing of up to three targeted gene disruptions (Gao et al., 2016b) and more recently five genomic loci were identified for gene integration (Schwartz et al., 2017b). As an alternative approach, similar gene disruptions have been successfully achieved using T7-transcribed sgRNAs with editing efficiencies of up to 60% (Morse et al., 2018). These initial advances have sparked interest in using a deactivated Cas9 (dCas9) system for gene expression modification. To this end, four different repressors were used to establish a CRISPR interference (CRISPRi) system to repress gene expression in Y. lipolytica (Zhang et al., 2018) that efficiently repressed expression of GFP as well as multiplexed repression of a 3-gene pathway that creates a violet pigment (Zhang et al., 2018). Likewise, a descriptive protocol for genome editing and repression with CRISPR-Cas9 and –dCas9 has been recently published
17
(Schwartz and Wheeldon, 2018). Finally, a VPR-dCas9 fusion was used to activate transcription of hrGFP and two native β-glucosidase genes (with multiplexed activation) demonstrating CRISPR activation (CRISPRa) (Schwartz et al., 2018). Collectively, these works demonstrate the wide utility of CRISPR systems in this host.
4.3.2 Transposases demonstrate unique potential for metabolic engineering Transposable elements (both native and heterologous) can be used for metabolic engineering applications. A diverse set of native transposable elements belonging to the retrotransposon (class I) and DNA transposon (class II) families have been identified in different strains of Y. lipolytica (Casaregola et al., 2000) including Ylt1, Tyl3 and Tyl6, belonging to the long terminal repeats (LTR or Zeta) group as well as Ylli from the non-LTR group (Casaregola et al., 2000; Magnan et al., 2016). An example of a DNA transposon is the Mutator-Like DNA transposon, Mutyl and Fotyl elements (Casaregola and Barth, 2013; Casaregola et al., 2000; Magnan et al., 2016; Neuvéglise et al., 2005). In some cases, these transposons may occur multiple times in the genome as is the case with Ylt1 which has 27 copies (partial and/or complete) in the genome of Y. lipolytica CLIB122 (Magnan et al., 2016). Such repetitive transposon systems can be used for targeting and amplifying copy number of transgenes (Juretzek et al., 2001; Le Dall et al., 1994; Madzak and Beckerich, 2013; Nicaud et al., 2003; Nicaud et al., 2002; Pignède et al., 2000; Schmid-Berger et al., 1994). As an example, a recent study used a DNA cut-and-paste transposon to amplify the copy number of a rate-limited polyketide synthase in Y. lipolytica (Yu et al., 2018). In addition to being useful for enhancing copy number, transposases (especially heterologous) can be used to create insertional mutagenesis libraries. One recent study
18
demonstrated an adaption of the piggyBac transposon system that is commonly used in eukaryotic cells and established effective generation of genome-wide mutagenesis libraries and scarless genomic modifications in Y. lipolytica (Wagner et al., 2018b). An additional functional genomics study created a saturated mutagenesis library of the entire genome using a Hermes DNA transposon allowing for identification of essential genes, evaluation of two genome-scale models, and selection of high-lipid strains (Patterson et al., 2018). Collectively, these studies demonstrate the unique utility of transposable elements for metabolic engineering in this yeast.
4.4 Multi-omics data and computational models facilitate rational engineering in Y. lipolytica As a final set of tools for Y. lipolytica, computational approaches have been established to model metabolism. Initial advances have focused on using models to fundamentally understand and improve lipogenesis. One group used lipidomics, metabolomics, and RNA-seq to study nitrogen starvation, identifying transcriptional regulators and demonstrating that regulation of lipid formation occurs through regulation of amino acid biosynthesis (Kerkhoven et al., 2016). Further transcriptomic work identified and validated additional transcription factors regulating lipogenesis (Trebulle et al., 2017). The effects of nitrogen starvation were modeled through the development of two mathematical models for lipid production based on Monod and inhibition kinetics as well as the Droop quota model approach (Robles-Rodriguez et al., 2018). Here, researchers used experimental data for calibration and validation and developed interesting controls for carbon to nitrogen ratio (Robles-Rodriguez et al., 2018). Finally, a more comprehensive stoichiometric model containing 645 genes, 1083 metabolites, and 1471 reactions was compiled from older databases and coupled with two knockout algorithms, OptGeneKnock
19
and IdealKnock, and an overexpression algorithm (APGC) to identify targets for TAG accumulation (Wei et al., 2017). These initial studies were capable of identifying targets to improve lipid formation in Y. lipolytica and serve as a platform to build models for other potential metabolites. Beyond lipid accumulation, in silico models have been applied for lycopene production (Zhao et al., 2017) and dodecanedioic acid production (Mishra et al., 2018). Thus, the use of big data is improving the ability to use rational engineering strategies to further improve this host. In conclusion, the synthetic and systems biology tools discussed here have paved the way to production of a variety of different chemicals through metabolic engineering. In the sections that follow, we demonstrate the wide applicability of these tools (when coupled with native metabolic capacities) to create chemicals including lipids, organic acids, sugar alcohols, and nonnative acetyl-CoA derived molecules. In the future, improvements of these tools (and development of novel tools) will facilitate efficient engineering efforts in Y. lipolytica.
5. The oleaginous phenotype and engineered lipid production of Y. lipolytica As has been alluded to above, Y. lipolytica has a distinctive metabolism when compared with the model yeast S. cerevisiae. While much information can be gained through comparison of these two organisms (Paiva et al., 2004; Beopoulos et al., 2008; Beopoulos et al., 2009; Christen and Sauer, 2011), it is the differences between these two organisms that make Y. lipolytica a valuable production host for a wide range of chemicals. In particular, as an oleaginous yeast, this organism has unique capabilities to synthesize high levels of intracellular lipids—the focus of many metabolic engineering approaches.
20
5.1 Lipid metabolism Key to the oleaginous phenotype of Y. lipolytica is this organism’s ability to synthesize de novo high levels of triacylglycerides (TAG). In order to achieve high TAG accumulation, Y. lipolytica has high flux through the fatty acid (FA) precursor, acetyl-CoA. In particular, this organism has adapted to funnel >80% of its glycolytic flux to acetyl-CoA production through the activity of pyruvate dehydrogenase (PDH) (Morgunov et al. 2004). However, this reaction takes place in the mitochondria, whereas cytosolic acetyl-CoA is required for FA biosynthesis. As a result, Y. lipolytica utilizes a citrate-malate antiport (Evans et al., 1983) coupled to several related enzymes (citrate synthase, CS; ATP-citrate lyase, ACL; malate dehydrogenase, MDH) to formulate a cycle which interconverts between the two pools of acetyl-CoA. A full description of this transport cycle (Figure 3) can be found in Wasylenko et al., 2015. This shuttling of acetylCoA across the mitochondrial membrane directly competes with the activity of the citric acid (TCA) cycle, a problem that is resolved through nitrogen regulation (Marchal et al., 1977; Ratledge and Wynn, 2002). In particular, when the cells have access to nitrogen, high ATP demands from protein synthesis and DNA replication drive the TCA cycle to operate at high capacity, oxidizing most of the acetyl-CoA. Conversely, when nitrogen is depleted, cellular AMP levels decrease (Boulton and Ratledge, 1983), leading to a decline in isocitrate dehydrogenase (IDH) activity (Botham and Ratledge, 1979), impedance of TCA cycle flux, and accumulation of mitochondrial citrate. The excess citrate is then used to export acetyl-CoA out of the mitochondria. More recently, it has been reported that increased dissolved oxygen levels in Y. lipolytica cultures lead to both a decrease in IDH activity and an increase in citrate production (Bellou et al., 2014), corroborating the proposed mechanism of acetyl-CoA shuttling. This
21
method of cytosolic acetyl-CoA generation is distinct from that of non-oleaginous yeasts, which typically use the pyruvate dehydrogenase bypass pathway (Beopoulos et al., 2011). Once transported into the cytosol, acetyl-CoA serves as the basic two-carbon building block for FA elongation and ultimately lipid synthesis. The enzymatic steps of these two processes are similar to that of most eukaryotic organisms and are summarized in Figure 3. Several comprehensive reviews on these topics have been published (Ratledge and Wynn, 2002; Coleman and Lee, 2004; Athenstaedt and Daum, 2006; Tehlivets et al., 2007), we here present the points that are of particular interest to the engineering of Y. lipolytica. Acetyl-CoA carboxylase (ACC1) catalyzes the first step in FA synthesis, converting acetyl-CoA to malonylCoA and priming it for subsequent chemistry as the basic elongation unit: acetyl-CoA + HCO3 + ATP
malonyl-CoA + ADP + Pi
Overexpression of this enzyme in Y. lipolytica has been shown to enhance FA flux (Tai and Stephanopoulos, 2013) and hence this step has been proposed to be rate limiting in the overall FA biosynthesis scheme (Pfleger et al., 2015). FA synthesis is mediated by the fatty acid synthase (FAS) complex where the two subunits, FAS1 and FAS2, catalyze the following reaction with NADPH as the reducing cofactor: acetyl-CoA + n malonyl-CoA + 2n NADPH
Cn+2-acyl-CoA + n CO2 + 2n NADP + n
CoASH For Y. lipolytica, the chain length for naturally synthesized fatty acids is typically 16 or 18 carbons (i.e., n = 7 or 8), yet this pathway is highly regulated to maintain homeostasis (Masoro, 1965; Goodridge, 1972; Heath et al., 1996; Davis and Cronan, 2001). Several studies have sought to alleviate this inhibition through either sequestering FAs into lipids (Tai and Stephanopoulos, 2013) or increasing the proportion of unsaturated FAs (Qiao et al. 2015). Both
22
of these engineering methods have demonstrated success in increasing lipid production. Finally the synthesized FA molecules condense with glycerol-3-phosphate (glyc3p) to produce TAGs stored in the lipid body: 3 FA + glyc3p
TAGs
Availability of the FAS cofactor, NADPH, is crucial for lipid synthesis. Engineering redox cofactor availability has recently been shown to enhance lipid yields significantly (Qiao et al. 2017). Unlike most other oleaginous yeasts where NADPH is produced primarily through the activity of malic enzymes (Wynn et al., 1997; Zhang et al., 2007), many studies suggest that Y. lipolytica regenerates NADPH almost exclusively through the oxidative pentose phosphate pathway (PPP) (Wasylenko et al., 2015). Zhang et al. reported that overexpressing the native copy of malic enzyme in Y. lipolytica did not increase its lipid production capabilities and that the enzyme prefers NAD+ over NADP+ (Zhang et al., 2013). Metabolic flux analysis performed on Y. lipolytica suggests a tight correlation between oxidative PPP and lipogenic fluxes, with minimal contribution from other candidate NADPH generating enzymes (Wasylenko et al., 2015; Liu et al., 2016). Furthermore, studies that focused on enhancing the oxidative PPP flux through overexpression of related enzymes have shown an increase in lipid synthesis, further strengthening this argument (Silverman et al., 2016; Yuzbasheva et al., 2017b). In addition to fatty acid biosynthesis, production of the TAG backbone chemical, glyc3p is also key to the lipogenic properties of Y. lipolytica. The enzyme, glyc3p dehydrogenase (GPDH) catalyzes the formation of glyc3p from the glycolytic intermediate, dihydroxyacetone (DHAP) (Beopoulos et al., 2008) and one study (Theirry et al.) suggests that this glyc3p production is key to this organism’s oleaginous traits (Dulermo and Nicaud, 2011). Apart from glyc3p synthesis, recent experimental evidence has also pointed to a tight correlation between
23
amino acid synthesis and lipid synthesis, in particular leucine metabolism was shown to have the greatest affect on lipid accumulation (Kerkhoven et al., 2016; Kerkhoven et al., 2017). Finally, the mechanisms of lipid catabolism in Y. lipolytica are also key to understanding the lipid overaccumulation phenotype (Fickers et al., 2005). Degradation of TAGs requires initial mobilization into FAs through the action of two lipid body bound lipases, TGL3 and TGL4 (Beopoulos et al., 2011). The FAs can then be activated to acyl-CoAs by acyl-CoA synthetase and subsequently broken down into two-carbon units through beta-oxidation (Tenagy et al., 2015). Beta-oxidation of acyl-CoAs occurs in a cyclic manner involving four enzymatic steps: acyl-CoA oxidase (Pox1-6), hydratase and dehydrogenase (both performed by the multifunctional enzyme Mfe2), and 3-ketoacyl-CoA thiolase (POT1) (Berninger et al., 1993; Wang et al., 1999; Blazeck et al. 2014). The degradation of FAs is typically carried out in the peroxisome of Y. lipolytica (Dulermo and Nicaud, 2011; Blazeck et al., 2014).
5.2 Metabolic engineering for lipid production Based on these native traits and capacity, it is not surprising that Y. lipolytica has been a host organism for engineered accumulation of lipids, especially in the form of TAGs. A variety of approaches have been used to optimize lipid production and we discuss these here briefly (additionally, these strategies are highlighted in Table 1). As examples, Liu et al. used a rationally engineered strain of Y. lipolytica (Blazeck et al., 2014) and identified a mutant allele in the lipid regulator mga2 that further improved lipid titer to nearly 40 g/L with saturated cells containing upwards of 90% dry cell weight lipid (Liu et al., 2015a). In their recent paper, Xu et al. tested several different pathways for cytosolic acetyl-CoA production in order to promote lipid accumulation. From this study (complementing an ACC1 and DGA1 overexpression strain
24
(Tai and Stephanopoulos, 2013)), it was determined that heterologous expression of the S. cerevisiae peroxisomal carnitine acyltransferase led to the highest lipid accumulation, producing 66.4 g/L TAGs using minimal media bioreactor fermentation (Xu et al., 2016). Another strategy combined a knockout of the peroxisomal biogenesis factor, PEX10 with overexpression of the native genes encoding glucose-6-phosphate dehydrogenase and acyl-CoA binding protein yielding a cell with lipid content of 30% dry cell weight (Yuzbasheva et al., 2017b). In a similar fashion, a beta-oxidation knockout combined with overexpressions of DGA2 and GPD1 yielded lipid at 55% dry cell weight under nitrogen limited conditions (Lazar et al., 2014; Sagnak et al., 2018). A different rational engineering approach employed overexpression of native oxidation stress related genes as well as those from the model organisms E. coli and S. cerevisiae leading to the production of 72.7 g/L lipid (Xu et al., 2017). Another study produced lipids at a titer of 98.9 g/L through heterologous expression of glycolytic enzymes that produce NADPH rather than NADH, this cofactor can then be used to promote lipid production (Qiao et al., 2017). Finally, through a recent transcriptomic analysis, researchers have identified a key native regulatory gene, MHY1 (Wang et al., 2017) that can increase lipogenesis when deleted leading to a content of 43.1% dry cell weight. Efforts in the field have been made to expand beyond the TAG production and leverage this host for the direct production of fatty acids. For example, fatty acids can be produced through the expression of a hybrid fatty acid synthase fused to a truncated version of E. coli thioesterase. Using this approach with unsupplemented, minimal medium, it was possible to produce 9.67 g/L of free fatty acids (Xu et al., 2016). An alternative approach to stimulate free fatty acid production involves inhibition of glycerol metabolism and beta-oxidation through a triple knockout strategy. Following evolution to derepress growth and oleate resistance, this
25
strain was led to fatty acid secretion at levels of 2.03 g/L following test tube fermentation when combined with overexpression of acetyl-CoA carboxylase and dodecane supplementation (Yuzbasheva et al., 2017a). Further modified products including fatty acid methyl esters have been produced. Production of 142.3 mg/L fatty acid ethyl esters was achieved through the combined expression of Acinetobacter baylyi wax-ester synthase with the lipogenic S. cerevisiae peroxisomal carnitine acyltransferase (Xu et al., 2016). Finally, wholesale shifts in the profile of fatty acids to long chain (C16 and C18) dicarboxylic acids at titers of 3.49 g/L (Abghari et al., 2017) have been achieved via the deletion of degradation pathways and coupled overexpression of omega-oxidation pathway genes. One of the larger, well-known rewiring of lipid production in Y. lipolytica is the engineering by DuPont to produce the nonnative fatty acid, omega-3-eicosapentaenoic acid (EPA), the main nutrient in fish oil. This effort involved introducing multiple heterologous copies of the Δ9-elongase and Δ8, Δ5 and Δ17-desaturase genes from a variety of algae and oomycete species (Xie et al., 2015). The combination of these complementations along with modification of native genes including overexpression of a C16/18 elongase gene and Δ12desaturase gene as well reduction of peroxisome biogenesis through a pex10 modification, led to a final strain able to accumulate EPA at 25% dry cell weight. This final strain is used to produce the New Harvest™ fish oil supplement as well as the source of feed for the EPA rich product, Verlasso® salmon. In similar fashion, Y. lipolytica has been engineered for the production of other nonnative fatty acid products including ricinoleic acid. This valuable product has been produced through the addition of a nonnative ∆12 hydroxylase gene in an engineered strain leading to ricinoleic acid production at 43% of total lipid content (Beopoulos et al., 2014). It is important to
26
note that this production was aided by supplementation with 2% ricinoleic acid to stimulate production. Ricinoleic acid from castor oil and other sources has likewise been fed to Y. lipolytica for bioconversion into gamma-decalactone where it has reached as high as 9.4 g/L titers depending on medium and fermentation conditions (Braga and Belo, 2014; Pagot et al., 1997; Wache et al., 1998). Another specialty fatty acid, gamma-linoleic acid has been produced at titers of 71.6 mg/L through the heterologous expression of a nonnative desaturase gene (Sun et al., 2017). The production of conjugated fatty acids (specifically conjugated linoleic acid) has been produced with titers of 302 mg/L through complementation of a linoleic acid isomerase along with reduced TAG formation and soybean oil feeding (Imatoukene et al., 2017). Finally, the production of arachidonic acid has been achieved (albeit at only 0.4% of total fatty acids) through a similar strategy (Liu et al., 2017a). In addition to these fatty acid products, the fuel hydrocarbon, pentane, has also been produced in the lipid accumulation MFE1 knockout background through heterologous expression of a soybean lipoxygenase enzyme (Blazeck et al., 2013a). Collectively, these studies show the flexibility and malleability of the lipid biosynthetic pathways in Yarrowia.
6. Recent advances in metabolic engineering Both industrial and lab-scale productions of chemicals beyond lipids and fatty acids have been explored for Y. lipolytica. In this section, we highlight the metabolic engineering efforts and feats that have been accomplished in this organism to expand the overall chemical profile of this promising host. In addition, to lower the industrial fermentation cost alternative carbon sources have been investigated through the metabolic engineering of catabolic pathways. These efforts are further summarized in Table 1.
27
6.1 Organic acids derived from the TCA cycle The overall oleaginous nature of Y. lipolytica and its metabolism as described above also means that this host has a high potential to produce TCA cycle intermediates. This ability is most pronounced in industrial production of citric acid using Y. lipolytica. In recent work (Kamzolova and Morgunov, 2017), a screening of 43 different wild isolates of different yeast species was evaluated for citric acid production. The best isolate belonged to Y. lipolytica and produced 85 g/L of citric acid in minimal medium with a yield of 0.70 g/g. Another recent study of note focused on transcriptome and fluxome characterization in a citrate producing strain ACA DC 50109 (Sabra et al., 2017). In addition to providing a valuable data set, this work was able to improve citrate production leading to 55 g/L from glucose alone and 50 g/L when using a dual substrate medium containing glycerol. Beyond citrate, this organism has been explored for the production of other related organic acids such as succinic acid, alpha-keto glutaric acid, and pyruvic acid. For example, an initial strategy was used to pair a high alpha-ketoglutaric acid producer with a two-step process whereby the alpha-ketoglutaric acid was then converted to succinic acid through a chemical hydrogen peroxide treatment (Kamzolova et al., 2009). More recent work has established a purely biological route to succinic aid through deletion of succinate dehydrogenase and CoAtransferase genes and overexpression of TCA cycle genes to enable 110.7 g/L of succinic acid production (Cui et al., 2017). However, these initial fermentations are limited by a compromised glucose consumption and thus glycerol was used as the carbon source. In order to enable production of succinic acid from fruit and vegetable waste hydrolysate (a cheap and glucose-rich material) a succinate dehydrogenase knockout strain was evolved for restored glucose
28
consumption, leading to production of 140.6 g/L of succinic acid (Li et al., 2018; Yang et al., 2017). In a similar manner, pyruvic and alpha-keto-glutaric acids have also been produced through the selection of a wild isolate followed by random mutagenesis and medium optimization leading to the production of 67.4 g/L alpha-keto glutaric acid and 39.1 g/L pyruvic acid (Zeng et al., 2015; Zeng et al., 2016; Zeng et al., 2017). Additionally, a thiamine auxotrophy was found to improve pyruvic acid production leading to production of 61.3 g/L (Morgunov et al., 2004). This work has been expanded to include the production of the nonnative, organic acid itaconic acid. This compound is a valuable platform chemical that can be converted to monomers for a variety of petroleum-derived products, including plastic monomers. Itaconic acid production in Y. lipolytica was enabled through heterologous expression of the Aspergillus terreus cis-aconitic acid decarboxylase. Production was further improved through cytosolic localization of the upstream, native aconitase enzyme, leading production of 4.6 g/L in bioreactor fermentation (Blazeck et al., 2015). Collectively, these results demonstrate the promise of using Y. lipolytica as a host for organic acid production.
6.2 Sugar products There is a high interest in the production of biorenewable sugar alcohols for use as food sweetners. In this regard, Y. lipolytica has been used in fermentations due to its capacity to readily produce erythritol. Substantial erythritol titers (170 g/L) were achieved through the identification of an isolate that does not produce citric acid as an overflow product when fermenting crude glycerol (Rymowicz et al., 2009). A bottom up approach to engineering involving overexpression of the pentose phosphate pathway transketolase gene or a predicted
29
erythrose reductase led to more modest titers (62.5 g/L and 78.1 g/L respectively) (Janek et al., 2017; Mirończuk et al., 2017). This pathway has also been exploited for the bioconversion of erythritol into the valuable product erythrulose, a valuable precursor to bioactive compounds including glyceraldehyde acetonide. Through overexpression of erythritol dehydrogenase and knockout of erythrolose kinase, this bioconversion yield reached 0.64 g/g (Carly et al., 2017; Trassaert et al., 2017).
6.3 Nonnative, non-lipid products derived from high acetyl-CoA flux The oleaginous nature of Y. lipolytica effectively stems from a high flux through acetylCoA and malonyl-CoA that can be diverted into an array of valuable compounds. The production of terpenoids is typically achieved through overexpression of the key native mevalonate pathway genes HMG1 and/or GGS1. When these strains are combined with heterologous pathways, modest titers of terpenoids have been produced. The complementation of phytoene synthase, phytoene dehydrogenase, and lycopene cyclase through expression of CarB and the bifunctional CarRP genes from Mucor circinelloides led to 2.22 mg/g dry cell weight beta-carotene (Gao et al., 2014). A multi-functional gene, carS from Schizochytrium sp. that encodes for all three functions led to 0.41mg/g dry cell weight (Gao et al., 2016a; Gao et al., 2014; Gao et al., 2017a). Further engineering efforts in these strains including overexpression of the mevalonate pathway gene ERG10 and knockout of associated beta-oxidation pathways led to the production of 4 g/L beta-carotene (Gao et al., 2017b). In another study, multiple copies of the biosynthetic genes (carB, carRP, and GGS1) were complemented into a variety of lipid overproducing strains and screened for beta-carotene production (Larroude et al., 2018). This study successfully achieved 6.5 g/L beta-carotene production during a bioreactor fermentation.
30
In a similar fashion, lycopene production draws from the same mevalonate pathway and thus similar strategies have been employed for lycopene overproduction. For example, by complementing a synthesis pathway from Pantoea ananatis into a high lipid producing strain, researchers achieved 16mg/g dry cell weight lycopene production (Matthäus et al., 2014). Likewise, production of lycopene was enhanced through FBA guided media optimization leading to bioreactor production of 242 mg/L lycopene (Nambou et al., 2015). Finally, as a demonstration of a CRISPR-Cas9 mediated, markerless gene integration strategy, a strain ultimately producing 21.1 mg/g dry cell weight lycopene in bioreactor fermentation was established (Schwartz et al., 2017a; Schwartz et al., 2017b). Beyond carotenoids, other terpenoids of interest have been explored in Y. lipolytica including the production of a downstream carotenoid product, astaxanthin (used as a food and feed additive) through the introduction of an additional gene from Pantoea ananatis, resulting in 54.6 mg/L in microtiter plate fermentation (Kildegaard et al., 2017). In another study, the αfarnesene synthase gene was fused to ERG20, yielding 260mg/L of the biofuel precursor (Yang et al., 2016). Finally, the key fragrance molecule, linalool, has also been produced in Y. lipolytica through melovanate pathway overexpression and heterologous expression leading to 6.96 mg/L linalool production (Cao et al., 2017). A variety of additional non-lipid and non-terpenoid products have been produced in Y. lipolytica. In a proof of concept paper describing a YaliBricks assembly technique, the pathway for violacein biosynthesis from Chromobacetrium violaceum was incorporated into Y. lipolytica to yield 31.5 mg/L production of this antibiotic pigment (Wong et al., 2017). Riboflavin overproduction was demonstrated in conjunction with a droplet-based sorting approach for secreted products (Wagner et al., 2018a). Specifically, this work led to 96 mg/L of riboflavin
31
after overexpression of RIB1 and RIB3 as well as knockout of beta-oxidation and subsequent EMS mutagenesis and droplet selection for an improved secretion phenotype (Wagner et al., 2018a). On the biopolymer side, the production pathway for poly-3-hydroxybutyrate from Ralstonia eutropha was incorporated into a strain of Y. lipolytica to result in nearly 7.35 g/L titers of this biodegradable plastic (Li et al., 2017). Finally, through the extensive rewiring of native metabolism along with the introduction of the Gerbera hybrida gene, g2ps1 (Markham et al., 2018), high level production of the polyketide, triacetic acid lactone was produced in Y. lipolytica. Specifically, overexpression of the pyruvate bypass pathway and the peroxisome biogenesis factor, PEX10 both allowed TAL production to become uncoupled from lipid accumulation. These results led to nearly 36 g/L of this polyketide, which can serve as a platform molecule and plastics precursor (Markham et al., 2018). In total, these studies all demonstrate the potential of rewiring the innate flux of acetyl-CoA and malonyl-CoA away from lipids and toward novel products.
6.4 Metabolic engineering to expand carbon source utilization To complement the product engineering approaches above and enable more biosustainable production, unique carbon sources have been explored for Y. lipolytica. For example, a variety of waste media sources have been tested for lipid production. In this regard, lipids can be produced from a wide variety of hydrophobic and hydrophilic carbon sources (Ledesma-Amaro and Nicaud, 2016) as well as waste carbon sources both glucose rich substrates such as various vegetable hydrolysates (Niehus et al., 2018; Quarterman et al., 2017) and oil rich substrates such as waste cooking oil (Katre et al., 2017). Human urine has also been explored as a novel, waste nitrogen source (Brabender et al., 2018).
32
Overall, hexose sugars such as glucose and fructose support rapid and robust growth of Y. lipolytica (Qiao et al., 2017; Lazar et al., 2014). Like many other yeasts, glucose is the preferred substrate and inhibits the uptake of most other carbon sources (Petit and Gancedo, 1999; Fickers et al., 2005). However, high concentrations of glucose (>300 g/L) impairs growth, presumably due to osmotic stress, thus requiring fed-batch cultures to achieve high product final titers (Qiao et al., 2015). Growth on other sugar-based components (like molasses) has been enabled by the expression of invertase activity such as through expression of S. cerevisiae SUC2 (Lazar et al., 2011; Hong et al., 2012; Lazar et al., 2013). In such strains, products including lipids and citric acid have been synthesized on sucrose or molasses as the sole carbon source (Moeller et al., 2013; Rakicka et al., 2015; Hapeta et al., 2017). Apart from sugars, Y. lipolytica has been extensively studied for its conversion of glycerol and crude glycerol obtained directly from industrial waste streams (Papanikolaou et al., 2002; Papanikolaou and Aggelis, 2002; Rymowicz et al., 2008; Rywińska et al., 2013). Catabolism of glycerol proceeds primarily through the “G3P pathway”, involving glycerol kinase (GK) encoded by GUT1 and glyc3p dehydrogenase (G3PDH) encoded by GUT2 (Dulermo and Nicaud, 2011; Mironczuk et al., 2016). Surprisingly, glycerol is one of the most preferred carbon sources for Y. lipolytica as it can repress the uptake of many other substrates (which glucose cannot), and in some cases even glucose (Mori et al., 2013; Sabra et al., 2017). This characteristic distinguishes Y. lipolytica from many other yeasts that regard glycerol as a nonfermentative carbon source. Y. lipolytica also has the distinctive feature of using hydrophobic substrates including alkanes and lipids. Once inside the cell, alkanes can be oxidized by a cytochrome P450dependent alkane monooxygenase system (Fickers et al., 2005) prior to entering the beta-
33
oxidation pathway. Likewise, the use of volatile fatty acids (VFAs) (such as acetic, propionic, and butyric acid) as a carbon source has been explored (Lian et al., 2012; Jiang et al., 2013; Pfleger, 2016). Owing to membrane diffusion of VFAs in the acidic form, a properly monitored pH can enabled continued uptake (Liu et al., 2016). Final efforts have been made to improve utilization of media components in ligocellulosic materials. For example, utilization of xylose was enabled by the addition of xylose reductase (XYR), xylitol dehydrogenase (XDH), and xylulose kinase (XYK) (Li and Alper, 2016). A blend of native an heterologous enzymes in this pathway have been explored (Li and Alper, 2016; Rodriguez et al., 2016). Other engineering efforts have enabled the expression of βglucosidases to enable direct growth on cellobiose (Guo et al., 2015). Additionally, the introduction of a cellodextrin transporter and an intracellular β-glucosidase both from Neurospora crassa can enable direct production of citric acid from cellobiose (Lane et al., 2015). Finally, recent advances have enabled the co-expression of six cellulolytic enzymes to create a pathway for simultaneous saccharification and fermentation directly from cellulose (Guo et al., 2017). By providing both a rapidly expanding product palette, as well as the ability to consume cheap waste products, Y. lipolytica allows for economical and biorenewable production of a wide range of products.
7. Perspectives The oleaginous industrial workhorse, Y. lipolytica, has long been observed to accumulate high levels of intracellular lipids. The proliferation of efficient genetic tools, screening systems and informative models has allowed metabolic engineers to not only improve overall lipid yields in this organism, but also to expand production to alternative, specialized lipid products. Y.
34
lipolytica has a robust history as an industrial producer of citric acid due to its innate ability to produce high levels of TCA cycle intermediates, and engineering efforts have likewise expanded and rerouted this capacity. Building upon this history, many recent studies demonstrate the ability of Y. lipolytica to serve as an ideal host for the production of heterologous secondary metabolites derived from acyl-CoA precursors. In addition to unique biochemistry and native physiology, Y. lipolytica lends itself to facile consumption of many waste products, thus providing access to diverse carbon sources and increasing the economic viability of industrial processes based on this host. The metabolic engineering work reviewed here demonstrates that Y. lipolytica is an attractive industrial host. However, there are many challenges left to be solved to harness the full potential of this lipogenic yeast. Looking to the future, further investment in expanding the genetic tools available for the rapid and efficient engineering of this host is necessary. In particular, stable episomal plasmids will allow for rapid genotype testing and enable researchers to leverage advances in protein engineering and rapid genetics. Although CRISPR technology recently showed high efficiencies in genome editing at a number of chromosomal loci in Y. lipolytica, some improvements of this technology are necessary to enable genome-wide efficiency. From a systems biology point of view, increased multi-omics data collection and an enhanced genome annotation will likewise yield useful databases and insightful metabolic models. With these improved technical and informational resources, it is anticipated that researchers will continue to expand beyond the view of Y. lipolytica as cell factory for lipids to become a cell factory for added-value lipid-derivatives as well as for diverse heterologous specialty chemicals. These limitations notwithstanding, the exciting research of the past several years provides a strong foundation for future work; with the application of emerging tools and
35
systems biology knowledge researchers will be able to unlock the potential of Y. lipolytica to serve as an industrial powerhouse. Acknowledgements This work was funded through the Office of Naval Research (ONR) under grant N00014-15-12785 (to H.A.), the Welch Foundation under grant F-1753 (to H.A.) and the Department of Energy under grant DE-SC0008744 (to G.S.). A. M. A. was funded by a postdoctoral fellowship from the Natural Sciences and Engineering Research Council of Canada (NSERC) under fellowship number PDF-488195-2016, and partly by the US DoE grant DE-SC0008744 mentioned above. We also thank the Koch Institute Swanson Biotechnology Center for technical support, specifically the Microscopy Core. Figure 1. Macroscopic appearance of Y. lipolytica colonies. Colonies of Y. lipolytica Po1f strain grown on (A) yeast-peptone-glucose (YPD) solid medium at 30 °C for 4 days, showing the convoluted white surface, (B) YPD at 30 °C for 2 days, showing the pale matte surface (C) YPD at 30 °C for 2 days then kept at 4 °C for 1 week, showing the smooth surface.
Figure 2. Lipid bodies of Y. lipolytica visualized using fluorescence microscopy. Cells were cultivated in YNB media grown under high carbon to nitrogen ratio. Cells were then stained using Nile Red stain and imaged by DIC and epifluorescence with a Nikon TE2000 microscope, equipped with a Lumencor, Spectra-CS, a 100 X, NA 1.49 oil objective, a red filter excitation /emission cube 530-560/590-650 (exposure time of 4 ms), an Andor Zyla camera. Lipid bodies appear as the red spots inside the cells.
36
Figure 3. Overview of the primary metabolic network in Y. lipolytica. Each color represents a different pathway: blue, glycolysis; green, pentose phosphate pathway; orange, TCA cycle; light red, acetyl-CoA shuttle and anaplerotic pathways; yellow, TAG synthesis pathways; dark red, lipid degradation pathways; black, substrate assimilation pathways; grey, major product synthesis pathways. Pathway localization with respect to specific subcellular organelles (represented by purple dotted squares) are also depicted. Arrow thickness in central carbon metabolism (glycolysis, TCA, and PPP) as well as TAG synthesis pathways represent relative pathway flux observed in a WT strain (Wasylenko et al. 2015). Table 1: Summary of many recent genetic strategies for native and nonnative chemical production in Y. lipolitca. For simplicity regulatory elements and copy number information have been removed. Genomic knockouts are represented by Δ followed by lowercase letters. In contrast desaturase and elongase genes are represented through the following notation: Δ-#-D or Δ-#-E. Native overexpressions are represented as all uppercase. All other notation refers heterologous genes or mutant alleles, following the notation used by the referenced study.. Referenc es
Product
Mediu Scale m
Backgr ound
Genetic Modifica tions
Evoluti on
Titer
ribofla vin product ion
96 mg/L
Native Products (Wagner et al., 2018a)
riboflavin
define d
flask
PO1f
Δpex10, URA3, RIB1, RIB3
(Gajdoš et al., 2017)
fatty acid
compl ex, glycer ol
flask
W29
Δdga1, Δdga2, Δlro1, Δare1, DGA2
37
9.9g/L
Yield
Rate
(Qiao et al., 2017)
fatty acid methyl esters
compl ex
biore actor
PO1g
LEU2, ACC1, DGA1, URA3, MCE, GapC
98.9 g/L
(Wang et al., 2016)
fatty alcohol
minim al
biore actor
PO1f
Tafar1, Δdga1, Δfao1
690.21 mg/L
(Yuzbash eva et al., 2017a)
free fatty acid minim al, glycer ol and dodec ane
flask
W29
Δgpd1, Δgut2, Δpex10, ACC1
(Xu et al., 2016)
free fatty acids
minim al
biore actor
PO1g
hybrid FAS1EcTesAtr uncated
9.67 g/L
(Abghari et al., 2017)
long chain dicarboxylic acids (16 and 18)
define d, glycer ol
biore actor
H222
Δpox1-6, Δsnf1, Δfaa1, ALK5, CPR, FAO1, ALK5
3.49 g/L
(Hanko et al., 2018)
long methyl ketones
compl ex
biore actor
PO1f
Δpot1, fadMSKL, fadBSKL, acoSKL
314.8 mg/L
(Blazeck et al., 2014)
triaceylglycer ides
minim al
biore actor
PO1f
Δpex10, Δmfe1, LEU2, URA3, DGA1
25.3g/ L
(Brabend er et al., 2018)
triaceylglycer ides
define d, urine
flask
PO1f
LEU2
0.653 g/L
38
glycero l derepre ssion, oleate resistan ce
1.3 g/L/h
0.018 g/g
2.03 g/L
0.06g/ g
0.04g/ L*h
(Katre et al., 2017)
Triaceylglyce rides
compl ex, waste cookin g oil
flask
NCIM 3589
random mutagene sis, selected for lipid productio n
(Liu et al., 2015b)
triaceylglycer ides
minim al
biore actor
PO1f
Δpex10, Δmfe1, LEU2, URA3, DGA1
(Liu et al., 2015a)
triaceylglycer ides
minim al
biore actor
PO1f
high lipid random mutant, DGA1, URA3, LEU2
25g/L
(Niehus et al., 2018)
triaceylglycer ides
agave bagass e hydrol ysate
biore actor
PO1d
Δpox1-6, Δtgl4, GDP1, DGA2, XR, XDH, XK, AnXPKA, AnACK
16.5g/ L
(Qiao et al., 2015)
triaceylglycer ides
compl ex
biore actor
PO1f
URA3, ACC1, DGA1, SCD
55g/L
(Quarter man et al., 2017)
triaceylglycer ides
switch grass hydrol ysate
flask
YB-420
(Lazar et al., 2014; Sagnak et al., 2018)
triaceylglycer ides
minera biore l actor
39
PO1f
Δtgl4, Δpox1–6, LEU2, DGA2, URA3, GPD1
67% DCW
random mutage nesis, selecte d for lipid content
39.1g/ L
0.56g/l /h
3.44g/ g sugars
1.85g/ L/h
1g/l/h
37.9 g/L
0.152 g/g
55% DCW
0.14g/ g
(Tai and Stephano poulos, 2013)
triaceylglycer ides
compl ex
biore actor
PO1f
URA3, ACC1, DGA1
61.4% DCW
(Wang et al., 2017)
triaceylglycer ides
compl ex
flask
ACADC 50109
Δmhy1
43.1% DCW
(Xu et al., 2016)
triaceylglycer ides
minim al
biore actor
PO1g
ACC1, DGA1, ScperCat 2
(Xu et al., 2017)
triaceylglycer ides
minim al
biore actor
PO1g
(Yuzbash eva et al., 2017b)
triaceylglycer ides
minim al
flask
W29
(Kamzol ova and Morguno v, 2017)
citric acid
minera biore l actor
VKM Y-2373
85 g/L
(Sabra et al., 2017)
citric acid
compl ex
biore actor
ACA DC 50109
55 g/L
(Morgun ov et al., 2004)
pyruvic acid
minim al, glycer ol
biore actor
VKM374/4
61.3 g/L
(Cui et al., 2017)
succinic acid
compl ex, glycer ol
biore actor
PO1f
Δsdh5, Δach1 ScPCK SCS2
110.7 g/L
(Gao et al., 2016b)
succinic acid
compl ex, crude glycer ol
biore actor
PO1f
∆sdh5, URA3
160g/L
40
0.195 g/g
0.143g /L/h
66.4 g/L
0.229 g/g
0.565 g/L/h
ACC1, DGA1, URA3, EcAldH, ScZWF, GSR, GPO
72.7 g/L
0.252 g/g
0.97 g/L/h
Δpex10, ZWF1, ACBP
30% DCW 0.70 g/g
0.71g/ g
76.3m g/g DCW/ h
(Kamzol ova et al., 2009)
succinic acid
minim al, ethano l
biore actor
VKM Y-2412
(Li et al., 2018; Yang et al., 2017)
succinic acid
fruit and vegeta ble waste hydrol ysate
biore actor
PO1f
∆sdh5, URA3
(Guo et al., 2014)
α keto glutaric acid
minim al, glycer ol
biore actor
WSHZ06
PDA1
43.3g/ L
(Yovkov a et al., 2014)
α keto glutaric acid
crude glycer ol
biore actor
H355
PYC1, IDP1
186g/L
(Zhou et al., 2012)
α keto glutaric acid
minim al, glycer ol
biore actor
WSHZ06
MmACL
56.5g/ L
(Zeng et al., 2015; Zeng et al., 2016; Zeng et al., 2017)
α keto glutaric acid and pyruvic acid
minim al, glycer ol
biore actor
WSHZ06
random mutagene sis, screened for low pH
67.4 g/L α keto glutari c acid, 39.1 g/L pyruvi c acid
(Janek et al., 2017)
erythritol
compl ex, glycer ol
biore actor
A101
ER
78.1 g/L
1 g/L/h
(Mirończ uk et al., 2017)
erythritol
compl ex, glycer ol
biore actor
MK1
TKL1
62.5 g/L
0.62g/ Lh
(Rymowi cz et al., 2009)
erythritol
crude glycer ol
biore actor
Wratisl avia K1
41
63.4 g/L
restore d glucose consum ption
58% EtOH fed
140.6 g/L
170g/L
0.69 g/L/h
3.7g/g
0.71 g/g
56%
0.74 g/L/h
1g/L/h
(Carly et al., 2018; Carly et al., 2017)
erythrulose
compl ex, erythri tol
biore actor
(Rakicka et al., 2017)
polyols
molass biore es, actor crude glycer ol
PO1d
Δeyk1, EYD1, URA3
Wratisl avia K1
Δura3, ScSUC2, GUT1
82.2 g/L erythri tol, 7.5 g/L arabito l, 11 g/L mannit ol
0.64 g/g
0.116g /g DCW* h
1.09 g/g
0.67g/ L/h
Nonnative Products (Wong et al., 2017)
violacein
define d
flask
W29
(Braga and Belo, 2014)
gammadecalactone
compl ex, castor oil
biore actor
W29
(Wache et al., 1998)
gammadecalactone
compl flask ex, methyl ricinol eate
PO1d
(Pagot et al., 1997)
gammadecalactone
compl flask ex, methyl ricinol eate
PO1d
(Liu et al., 2017b)
arachidonic acid
compl ex
PO1f
42
flask
vioA, vioB, vioC, vioD, vioE
31.5 mg/L
5.4 g/L
Δaco3
195 mg/L
9.4g/L
URA3, Δ6-D, Δ-6E, Δ-5-D
215 mg/L/ h
0.42% total FA
0.58g/ g methy l ricino leate
13.6m g/g/h
(Imatouk ene et al., 2017)
conjugated linoleic acids
compl biore ex, actor soybea n oil
PO1d
Δpox1-6, Δdga1, Δdga2, Δlro1, Δfad2, FAD2, oPAI
302mg /L
(Imatouk ene et al., 2017)
conjugated linoleic acids
compl ex
biore actor
PO1d
Δpox1-6, Δdga1, Δdga2, Δlro1, Δfad2, FAD2, oPAI
180 mg/L
(Xu et al., 2016)
fatty acid ethyl esters
minim al
flask
PO1g
AbAtfA, ScperCat 2
142.5 mg/L
(Xu et al., 2016)
fatty alcohols
minim al
biore actor
PO1g
Maqu222 0, EcfadD
2.15 g/L
(Xu et al., 2016)
fatty alkanes
minim al
flask
PO1g
MmCAR, BsuSfp, PmADO
23.3 mg/L
(Sun et al., 2017)
gammalinoleic acid
compl ex
flask
PO1f
Δ-6-D
71.6 mg/L
(Gao et al., 2015)
Mediumchain-length polyhydroxya lkanoates
compl ex, triolei n
flask
PO1h
PhaC1 with PTS1 signal and Kozak sequence
1.11g/ L
(Xie et al., 2017)
omega-3 eicosapentae noic acid
compl ex
biore actor
ATCC 20362
Δpex10, Δ-9-E, Δ8-D, Δ-5D, Δ-17D, C1618-E, Δ12-D, acyltransf erase
25% biomas s
43
(Xie et al., 2015)
omega-3 eicosapentae noic acid
compl ex
biore actor
ATCC 20362
Δpex10, Δ-9-E, Δ8-D, Δ-5D, Δ-17D, C1618-E, Δ12-D, acyltransf erase
25% DCW
(Xue et al., 2013)
omega-3 eicosapentae noic acid
compl ex
shake flask
ATCC 20362
Δpex10, Δlip1, Δscp2, Δyali0c18 71, Δ-12D, Δ-9-E, C16/18E, Δ-8-D, Δ-5-D, Δ17-D
15% DCW
(Blazeck et al., 2013a)
pentane
define d
1L bottle s
po1f
Δmfe1, Gmlox1
4.98m g/L
(Beopoul os et al., 2014)
ricinoleic acid
compl ex, ricinol eic acid
tube
PO1d
Δpox1-6, Δdga1, Δdga2, Δlro1, ∆fad2, CpFAH1 2, YlLRO1
43% total lipid
63mg/ g DCW
(Blazeck et al., 2015)
itaconic acid
minim al
biore actor
PO1f
LEU2, URA3, AtCAD, ACOnoM LS
4.6g/L
0.058 g/g
(Li et al., 2017)
poly-3hydroxybutyr ate
compl biore ex, actor acetate
PO1g
URA3, phaC, phaA, phaB
7.35 g/L
44
0.045g /L/h
(Markha m et al., 2018)
triacetic acid lactone
compl biore ex, actor acetate spike
PO1f
g2ps1, LEU2, URA3, ACS1, ALD5, PDC2, ACC1
35.9 g/L
0.164 g/g*
(Markha m et al., 2018)
triacetic acid lactone
define d
tube
PO1f
g2ps1, LEU2, URA3, PEX10
4.1 g/L
0.203 g/g
(Yu et al., 2018)
triacetic acid lactone
minim al
biore actor
W29
URA3, LEU2, g2ps1
2.6g/L
13.80 %
(Zhang et al., 2017)
campesterol
compl ex
biore actor
ATCC 201249
Δerg5, DHCR7, POX2
942 mg/L
(Kildegaa rd et al., 2017)
astaxanthin
compl ex
micro titer plate
GB20
Δmus51, nugmHtg2, ndh2i, XdcrtI, XdcrtYB, XdcrtE, HMG1, PSQS1_50bp: SQS1, PscrtW, PacrtZ
54.6 mg/L
3.5 mg/g DCW
(Cao et al., 2017)
linalool
compl flask ex, citrate and pyruva te
PO1f
ERG20F88 W-N119W , HMG1, IDI1
6.96 mg/L
939 μg/g DCW
(Schwart z et al., 2017a; Schwartz et al., 2017b)
lycopene
compl ex
PO1f
HMG1, CrtE, CrtB, CrtI, LEU2, MVD1, ERG8
45
biore actor
21.1 mg/g DCW
max 0.21 g/L/h
(Nambou et al., 2015)
lycopene
compl ex
biore actor
PO1f
CrtE, CrtB, CrtI
(Matthäu s et al., 2014)
lycopene
minim al
biore actor
H222
ScSUC2, Δpox1-6, Δgut2, CrtB, CrtI, GGS1, HMG1
(Yang et al., 2016)
α-farnesene
compl ex, dodec ane
biore actor
PO1h
tHMG1, hybrid OptFSERG20, IDI
(Gao et al., 2016a; Gao et al., 2014; Gao et al., 2017a)
β-carotene
compl ex
flask
ATCC MYA2613
ScSUC2, Δku70, Δku80, GGS1, βcarS, URA3
0.41m g/g DCW
(Gao et al., 2014)
β-carotene
compl ex
flask
ATCC 201249
GGS1, CarB, CarRP, URA3
2.22 mg/g DCW
(Larroud e et al., 2018)
β-carotene
compl ex
biore actor
Po1D
DGA2, GPD1, Δpox1-6, Δtgl4, GGS1, CarB, CarRP, tHMG1
46
242 mg/L 16 mg/g CDW
259.98 mg/L
6.5 g/L
33.98 mg/g DCW
89.6 mg/g DCW
(Kildegaa rd et al., 2017)
β-carotene
compl ex
micro titer plate
GB20
Δmus51, nugmHtg2, ndh2i, XdcrtI, XdcrtYB, XdcrtE, HMG1, PSQS1_50bp: SQS1
797.1 mg/L
(Gao et al., 2017a)
β-carotene
compl ex
biore actor
ATCC MYA2613
ScSUC2 Δku70, CarB, CarRP Δpox4, ERG10
4 g/L
(Celińska and Grajek, 2013; Celińska et al., 2013)
2phenylethano l
minim al, glycer ol and L-Phe
biore actor
NCYC3 825
ScSUC2, dhaB, dhaB2, dhaT
1.98g/ L
0.014 g/L/h
0.31g/ g
20mg/ (Lxh)
References Abghari, A., Chen, S., 2014. Yarrowia lipolytica as an oleaginous cell factory platform for production of fatty acid-based biofuel and bioproducts. Frontiers in Energy Research. 2. Abghari, A., Madzak, C., Chen, S., 2017. Combinatorial engineering of Yarrowia lipolytica as a promising cell biorefinery platform for the de novo production of multi-purpose long chain dicarboxylic acids. Fermentation. 3, 40. Athenstaedt, K., 2010. Neutral lipids in yeast: Synthesis, storage and degradation. In: Timmis, K. N., (Ed.), Handbook of Hydrocarbon and Lipid Microbiology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 471-480. Athenstaedt, K., and Daum, G., 2006. The life cycle of neutral lipids: synthesis, storage and degradation. Cellular and Molecular Life Sciences. 63 (12), 1355–69. Athenstaedt, K., Jolivet, P., Boulard, C., Zivy, M., Negroni, L., Nicaud, J. M., Chardot, T., 2006. Lipid particle composition of the yeast Yarrowia lipolytica depends on the carbon source. Proteomics. 6, 1450-1459. Bakkaiova, J., Arata, K., Matsunobu, M., Ono, B., Aoki, T., Lajdova, D., Nebohacova, M., Nosek, J., Miyakawa, I., Tomaska, L., 2014. The strictly aerobic yeast Yarrowia lipolytica tolerates loss of a mitochondrial DNA-packaging protein. Eukaryotic Cell. 13, 1143-1157. Bankar, A. V., Kumar, A. R., Zinjarde, S. S., 2009. Environmental and industrial applications of Yarrowia lipolytica. Applied Microbiology and Biotechnology. 84, 847. 47
Barth, G., Gaillardin, C., 1996. Yarrowia lipolytica. Nonconventional Yeasts in Biotechnology: A Handbook. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 313-388. Barth, G., Gaillardin, C., 1997. Physiology and genetics of the dimorphic fungus Yarrowia lipolytica. FEMS Microbiology Reviews. 19, 219-237. Bellou, S., Makri, A., Triantaphyllidou, I.-E., Papanikolaou, S., Aggelis, G., 2014. Morphological and metabolic shifts of Yarrowia lipolytica induced by alteration of the dissolved oxygen concentration in the growth environment. Microbiology. 160, 807-817. Beopoulos, A., Mrozova, Z., Thevenieau, F., Le Dal, M.-T., Hapala, I., Papanikolaou, S., Chardot, T., Nicaud, J.-M., 2008. Control of lipid accumulation in the yeast Yarrowia lipolytica. Applied and Environmental Microbiology. 74, 7779-7789. Beopoulos, A., Chardot, T., Nicaud, J.-M., 2009. Yarrowia lipolytica: A model and a tool to understand the mechanisms implicated in lipid accumulation. Biochimie. 91, 692-696. Beopoulos, A., Nicaud, J.-M., and Gaillardin, C., 2011. An overview of lipid metabolism in yeasts and its impact on biotechnological processes. Applied Microbiology and Biotechnology 90(4), 1193–1206. Beopoulos, A., Nicaud, J.-M., 2012. Yeast: A new oil producer? OCL. 19, 22-28. Beopoulos, A., Verbeke, J., Bordes, F., Guicherd, M., Bressy, M., Marty, A., Nicaud, J.M., 2014. Metabolic engineering for ricinoleic acid production in the oleaginous yeast Yarrowia lipolytica. Applied Microbiology and Biotechnology. 98, 251-262. Berninger, G., Schmidtchen, R., Casel, G., Knorr, A., Rautenstrauss, Kanau, W.-H., Schweizer, E., 1993. Structure and metabolic control of the Yarrowia lipolytica peroxisomal 3‐ oxoacyl‐ CoA‐ thiolase gene. European Journal of Biochemistry. 216, 607–13. Blazeck, J., Hill, A., Jamoussi, M., Pan, A., Miller, J., Alper, H. S., 2015. Metabolic engineering of Yarrowia lipolytica for itaconic acid production. Metabolic engineering. 32, 6673. Blazeck, J., Hill, A., Liu, L., Knight, R., Miller, J., Pan, A., Otoupal, P., Alper, H. S., 2014. Harnessing Yarrowia lipolytica lipogenesis to create a platform for lipid and biofuel production. Nature communications. 5, 3131. Blazeck, J., Liu, L., Knight, R., Alper, H. S., 2013a. Heterologous production of pentane in the oleaginous yeast Yarrowia lipolytica. Journal of Biotechnology. 165, 184-194. Blazeck, J., Liu, L., Redden, H., Alper, H., 2011. Tuning gene expression in Yarrowia lipolytica by a hybrid promoter approach. Appl Environ Microbiol. 77, 7905-14. Blazeck, J., Reed, B., Garg, R., Gerstner, R., Pan, A., Agarwala, V., Alper, H. S., 2013b. Generalizing a hybrid synthetic promoter approach in Yarrowia lipolytica. Appl Microbiol Biotechnol. 97, 3037-52. Bordes, F., Fudalej, F., Dossat, V., Nicaud, J. M., Marty, A., 2007. A new recombinant protein expression system for high-throughput screening in the yeast Yarrowia lipolytica. Journal of microbiological methods. 70, 493-502. Botham, P. A., and Ratledge, C., 1979. A biochemical explanation for lipid accumulation in Candida 107 and other oleaginous micro-organisms. Journal of General Microbiology. 114, 361-75. Brabender, M., Shabbir-Hussain, M., Rodriguez, G., Blenner, M. A., 2018. Urea and urine are a viable and cost-effective nitrogen source for Yarrowia lipolytica biomass and lipid accumulation. Applied Microbiology and Biotechnology. 102, 2313-2322.
48
Braga, A., Belo, I., 2014. Production of gamma-decalactone by Yarrowia lipolytica: insights into experimental conditions and operating mode optimization. J Chem Technol Biotechnol. 90, 559-565. Bredeweg, E. L., Pomraning, K. R., Dai, Z., Nielsen, J., Kerkhoven, E. J., Baker, S. E., 2017. A molecular genetic toolbox for Yarrowia lipolytica. Biotechnology for Biofuels. 10, 2-22. Cao, X., Wei, L. J., Lin, J. Y., Hua, Q., 2017. Enhancing linalool production by engineering oleaginous yeast Yarrowia lipolytica. Bioresource Technology. 245, 1641-1644. Carly, F., Steels, S., Telek, S., Vandermies, M., Nicaud, J. M., Fickers, P., 2018. Identification and characterization of EYD1, encoding an erythritol dehydrogenase in Yarrowia lipolytica and its application to bioconvert erythritol into erythrulose. Bioresource Technology. 247, 963-969. Carly, F., Vandermies, M., Telek, S., Steels, S., Thomas, S., Nicaud, J. M., Fickers, P., 2017. Enhancing erythritol productivity in Yarrowia lipolytica using metabolic engineering. Metabolic Engineering. 42, 19-24. Casaregola, S., Barth, G., 2013. Transposable elements and their activities in Y. lipolytica. In: Barth, G., (Ed.), Yarrowia lipolytica: Genetics, Genomics, and Physiology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 57-77. Casaregola, S., Neuvéglise, C., Lépingle, A., Bon, E., Feynerol, C., Artiguenave, F., Wincker, P., Gaillardin, C., 2000. Genomic exploration of the hemiascomycetous yeasts: 17. Yarrowia lipolytica. FEBS Letters. 487, 95-100. Celińska, E., Grajek, W., 2013. A novel multigene expression construct for modification of glycerol metabolism in Yarrowia lipolytica. Microbial Cell Factories. 12, 102. Celińska, E., Kubiak, P., Białas, W., Dziadas, M., Grajek, W., 2013. Yarrowia lipolytica: the novel and promising 2-phenylethanol producer. Journal of Industrial Microbiology & Biotechnology. 40, 389-392. Celińska, E., Ledesma-Amaro, R., Larroude, M., Rossignol, T., Pauthenier, C., Nicaud, J. M., 2017. Golden Gate Assembly system dedicated to complex pathway manipulation in Yarrowia lipolytica. Microbial biotechnology. 10, 450-455. Chen, D. C., Beckerich, J. M., Gaillardin, C., 1997. One-step transformation of the dimorphic yeast Yarrowia lipolytica. Appl Microbiol Biotechnol. 48, 232-5. Christen, S., Sauer, U., 2011. Intracellular characterization of aerobic glucose metabolism in seven yeast species by 13C flux analysis and metabolomics. FEMS Yeast Research. 11, 263272. Coleman, R. A., and Lee, D. P., 2004. Enzymes of triacylglycerol synthesis and their regulation. Progress in Lipid Research. 43, 134–76. Cui, Z., Gao, C., Li, J., Hou, J., Lin, C. S. K., Qi, Q., 2017. Engineering of unconventional yeast Yarrowia lipolytica for efficient succinic acid production from glycerol at low pH. Metabolic Engineering. 42, 126-133. Curran, K. A., Morse, N. J., Markham, K. A., Wagman, A. M., Gupta, A., Alper, H. S., 2015. Short synthetic terminators for improved heterologous gene expression in yeast. ACS Synth Biol. 4, 824-32. Czabany, T., Wagner, A., Zweytick, D., Lohner, K., Leitner, E., Ingolic, E., Daum, G., 2008. Structural and biochemical properties of lipid particles from the yeast Saccharomyces cerevisiae. Journal of Biological Chemistry. 283, 17065-17074. Daley, J. M., Palmbos, P. L., Wu, D., Wilson, T. E., 2005. Nonhomologous end joining in yeast. Annual Review of Genetics. 39, 431-451. 49
Davidow, L. S., Apostolakos, D., O'Donnell, M. M., Proctor, A. R., Ogrydziak, D. M., Wing, R. A., Stasko, I., DeZeeuw, J. R., 1985. Integrative transformation of the yeast Yarrowia lipolytica. Current Genetics. 10, 39-48. Davis, M. S., and Cronan, J. E., 2001. Inhibition of Escherichia coli acetyl coenzyme A carboxylase by acyl-acyl carrier protein. Journal of Bacteriology. 183, 1499–1503. Domínguez, A., Deive, F. J., Sanromán, M. A., Longo, M. A., 2010. Biodegradation and utilization of waste cooking oil by Yarrowia lipolytica CECT 1240. European Journal of Lipid Science and Technology. 112, 1200-1208. Dujon, B., Sherman, D., Fischer, G., Durrens, P., Casaregola, S., Lafontaine, I., de Montigny, J., Marck, C., Neuveglise, C., Talla, E., Goffard, N., Frangeul, L., Aigle, M., Anthouard, V., Babour, A., Barbe, V., Barnay, S., Blanchin, S., Beckerich, J.-M., Beyne, E., Bleykasten, C., Boisrame, A., Boyer, J., Cattolico, L., Confanioleri, F., de Daruvar, A., Despons, L., Fabre, E., Fairhead, C., Ferry-Dumazet, H., Groppi, A., Hantraye, F., Hennequin, C., Jauniaux, N., Joyet, P., Kachouri, R., Kerrest, A., Koszul, R., Lemaire, M., Lesur, I., Ma, L., Muller, H., Nicaud, J.-M., Nikolski, M., Oztas, S., Ozier-Kalogeropoulos, O., Pellenz, S., Potier, S., Richard, G.-F., Straub, M.-L., Suleau, A., Swennen, D., Tekaia, F., Wesolowski-Louvel, M., Westhof, E., Wirth, B., Zeniou-Meyer, M., Zivanovic, I., Bolotin-Fukuhara, M., Thierry, A., Bouchier, C., Caudron, B., Scarpelli, C., Gaillardin, C., Weissenbach, J., Wincker, P., Souciet, J.-L., 2004. Genome evolution in yeasts. Nature. 430, 35-44. Dulermo, T., and Nicaud J.-M., 2011. Involvement of the G3P shuttle and beta-oxidation pathway in the control of TAG synthesis and lipid accumulation in Yarrowia lipolytica. Metabolic Engineering. 13, 482–91. Egermeier, M., Russmayer, H., Sauer, M., Marx, H., 2017. Metabolic flexibility of Yarrowia lipolytica growing on glycerol. Frontiers in Microbiology. 8. Esser, K., Stahl, U., 1976. Cytological and genetic studies of the life cycle of Saccharomycopsis lipolytica. Molecular and General Genetics MGG. 146, 101-106. Evans, C. T., Scragg, A. H., Ratledge, C., 1983. A comparative study of citrate efflux from mitochondria of oleaginous and mon-oleaginous yeasts. European Journal of Biochemistry. 130, 195-204. Fabre, E., Nicaud, J.-M., Lopez, M. C., Gaillardin, C., 1991. Role of the proregion in the production and secretion of the Yarrowia lipolytica alkaline extracellular protease. Journal of Biological Chemistry. 266, 3782-90. Fickers, P., Benetti, P. H., Waché, Y., Marty, A., Mauersberger, S., Smit, M. S., Nicaud, J. M., 2005. Hydrophobic substrate utilisation by the yeast Yarrowia lipolytica, and its potential applications. FEMS Yeast Research. 5, 527-543. Fickers, P., Le Dall, M. T., Gaillardin, C., Thonart, P., Nicaud, J. M., 2003. New disruption cassettes for rapid gene disruption and marker rescue in the yeast Yarrowia lipolytica. Journal of microbiological methods. 55, 727-37. Fickers, P., Nicaud, J.-M., Gaillardin, C., Destain, J., Thonart, P., 2004. Carbon and nitrogen sources modulate lipase production in the yeast Yarrowia lipolytica. Journal of Applied Microbiology. 96, 742-749. Fournier, P., Abbas, A., Chasles, M., Kudla, B., Ogrydziak, D. M., Yaver, D., Xuan, J. W., Peito, A., Ribet, A. M., Feynerol, C., 1993a. Colocalization of centromeric and replicative functions on autonomously replicating sequences isolated from the yeast Yarrowia lipolytica. Proceedings of the National Academy of Sciences. 90, 4912.
50
Fournier, P., Gaillardin, C., Kudla, B., Heslot, H., ARS sequence which is efficacious in Yarrowia lipolytica. Agronomique Inst Nat Rech, US, 1993b. Fournier, P., Guyaneux, L., Chasles, M., Gaillardin, C., 1991. Scarcity of ars sequences isolated in a morphogenesis mutant of the yeast Yarrowia lipolytica. Yeast. 7, 25-36. Gaillardin, C., Mekouar, M., Neuvéglise, C., 2013. Comparative genomics of Yarrowia lipolytica. In: Barth, G., (Ed.), Yarrowia lipolytica: Genetics, Genomics, and Physiology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 1-30. Gaillardin, C., Ribet, A. M., 1987. LEU2 directed expression of beta-galactosidase activity and phleomycin resistance in Yarrowia lipolytica. Curr Genet. 11, 369-75. Gajdoš, P., Nicaud, J.-M., Čertík, M., 2017. Glycerol conversion into a single cell oil by engineered Yarrowia lipolytica. Engineering in Life Sciences. 17, 325-332. Gammage, P. A., Moraes, C. T., Minczuk, M., 2017. Mitochondrial genome engineering: The revolution may not be CRISPR-Ized. Trends in Genetics. 34, 101-110. Gancedo, J. M., 2001. Control of pseudohyphae formation in Saccharomyces cerevisiae. FEMS Microbiology Reviews. 25, 107-123. Gao, C., Qi, Q., Madzak, C., Sze, C., Lin, K., 2015. Exploring medium ‑ chain ‑ length polyhydroxyalkanoates production in the engineered yeast Yarrowia lipolytica. Journal of Industrial Microbiology & Biotechnology. 42, 1255-1262. Gao, C., Yang, X., Wang, H., Rivero, C. P., Li, C., Cui, Z., Qi, Q., Lin, C. S. K., Sze, C., Lin, K., 2016a. Robust succinic acid production from crude glycerol using engineered Yarrowia lipolytica. Biotechnology for biofuels. 9, 179. Gao, S., Han, L., Zhu, L., Ge, M., 2014. One-step integration of multiple genes into the oleaginous yeast Yarrowia lipolytica. Biotechnology Journal. 36, 2523-2528. Gao, S., Tong, Y., Wen, Z., Zhu, L., Ge, M., Chen, D., Jiang, Y., Yang, S., 2016b. Multiplex gene editing of the Yarrowia lipolytica genome using the CRISPR-Cas9 system. Journal of Industrial Microbiology & Biotechnology. 43, 1085-1093. Gao, S., Tong, Y., Zhu, L., Ge, M., Jiang, Y., Chen, D., Yang, S., 2017a. Production of βcarotene by expressing a heterologous multifunctional carotene synthase in Yarrowia lipolytica. Biotechnology Letters. 39, 921-927. Gao, S., Tong, Y., Zhu, L., Ge, M., Zhang, Y., Chen, D., Jiang, Y., Yang, S., 2017b. Iterative integration of multiple-copy pathway genes in Yarrowia lipolytica for heterologous βcarotene production. Metabolic Engineering. 41, 192-201. Garay, L. A., Boundy-Mills, K. L., German, J. B., 2014. Accumulation of high-value lipids in single-cell microorganisms: A mechanistic approach and future perspectives. Journal of Agricultural and Food Chemistry. 62, 2709-2727. Gasmi, N., Ayed, A., Nicaud, J.-M., Kallel, H., 2011. Design of an efficient medium for heterologous protein production in Yarrowia lipolytica: case of human interferon alpha 2b. Microbial Cell Factories. 10, 38. Goodridge, A.G., 1972. Regulation of the activity of acetyl coenzyme A carboxylase by palmitoyl coenzyme A and citrate. Journal of Biological Chemistry. 247, 6946–52. Groenewald, M., Boekhout, T., Neuveglise, C., Gaillardin, C., van Dijck, P. W., Wyss, M., 2014. Yarrowia lipolytica: safety assessment of an oleaginous yeast with a great industrial potential. Critical Reviews in Microbioly. 40, 187-206. Guo, H., Madzak, C., Du, G., Zhou, J., Chen, J., 2014. Effects of pyruvate dehydrogenase subunits overexpression on the α-ketoglutarate production in Yarrowia lipolytica WSH-Z06. Applied Microbiology and Biotechnology. 98, 7003-7012. 51
Guo, Z., Duquesne, S., Bozonnet, S., Cioci, G., Nicaud, J.-M., Marty, A., and O’Donohue, M. J., 2015. Development of cellobiose-degrading ability in Yarrowia lipolytica strain by overexpression of endogenous genes. Biotechnology for Biofuels. 8, 1–16. Guo, Z., Duquesne, S., Bozonnet, S., Cioci, G., Nicaud, J.-M., Marty, A., O’Donohue, M.J., 2017. Conferring cellulose-degrading ability to Yarrowia lipolytica to facilitate a consolidated bioprocessing approach. Biotechnology for Biofuels 10, 132. Hanko, E. K. R., Denby, C. M., Nogué, S., Lin, W., Ramirez, K. J., Singer, C. A., Beckham, G. T., Jay, D., Lin, W., Ramirez, K. J., Singer, C. A., Beckham, G. T., Keasling, J. D., 2018. Engineering β-oxidation in Yarrowia lipolytica for methyl ketone production. Metabolic Engineering. 48, 52-62. Hapeta, P., Rakicka, M., Dulermo, R., Gamboa-Meléndez, H., Crutz-Le Coq, A. M., Nicaud, J.-M., Lazar, Z., 2017. Transforming sugars into fat-lipid biosynthesis using different sugars in Yarrowia lipolytica. Yeast 34, 293–304. Hassanshahian, M., Tebyanian, H., Cappello, S., 2012. Isolation and characterization of two crude oil-degrading yeast strains, Yarrowia lipolytica PG-20 and PG-32, from the Persian Gulf. Marine Pollution Bulletin. 64, 1386-1391. Heslot, H., Genetics and genetic engineering of the industrial yeast Yarrowia lipolytica. Springer Berlin Heidelberg, Berlin, Heidelberg, 1990, pp. 43-73. Holkenbrink, C., Dam, M. I., Kildegaard, K. R., Beder, J., Dahlin, J., Doménech Belda, D., Borodina, I., 2018. EasyCloneYALI: CRISPR/Cas9-based synthetic toolbox for engineering of the yeast Yarrowia lipolytica. Biotechnology Journal. 1700543-n/a. Hong, S. P., Seip, J., Walters‐Pollak, D., Rupert, R., Jackson, R., Xue, Z., Zhu, Q., 2012. Engineering Yarrowia lipolytica to express secretory invertase with strong FBA1IN promoter. Yeast. 29, 59-72. Hurtado, C. A. R., Beckerich, J.-M., Gaillardin, C., Rachubinski, R. A., 2000. A Rac homolog is required for induction of hyphal growth in the dimorphic yeast Yarrowia lipolytica. Journal of Bacteriology. 182, 2376-2386. Imatoukene, N., Verbeke, J., Beopoulos, A., Idrissi Taghki, A., Thomasset, B., Sarde, C. O., Nonus, M., Nicaud, J.-M., 2017. A metabolic engineering strategy for producing conjugated linoleic acids using the oleaginous yeast Yarrowia lipolytica. Applied Microbiology and Biotechnology. 101, 4605-4616. Isakova, E. P., Epova, E. Y., Sekova, V. Y., Trubnikova, E. V., Kudykina, Y. K., Zylkova, M. V., Guseva, M. A., Deryabina, Y. I., 2015. The engineering of a Yarrowia lipolytica yeast strain capable of homologous recombination of the mitochondrial genome. Prikladnaia biokhimiia i mikrobiologiia. 51, 319-25. Jacques, N., Casaregola, S., 2008. Safety assessment of dairy microorganisms: The hemiascomycetous yeasts. International Journal of Food Microbiology. 126, 321-326. Janek, T., Dobrowolski, A., Biegalska, A., Mirończuk, A. M., 2017. Characterization of erythrose reductase from Yarrowia lipolytica and its influence on erythritol synthesis. Microbial Cell Factories. 16, 1-13. Jiang, J., Zhang, Y., Li, K., Wang, Q., Gong, C., Li, M., 2013. Volatile fatty acids production from food waste: effects of pH, temperature, and organic loading rate. Bioresource Technology. 143, 525–30. Jost, B., Holz, M., Aurich, A., Barth, G., Bley, T., Müller, R. A., 2015. The influence of oxygen limitation for the production of succinic acid with recombinant strains of Yarrowia lipolytica. Applied Microbiology and Biotechnology. 99, 1675-1686. 52
Juretzek, T., Le Dall, M. T., Mauersberger, S., Gaillardin, C., Barth, G., Nicaud, J. M., 2001. Vectors for gene expression and amplification in the yeast Yarrowia lipolytica. Yeast. 18, 97-113. Kamzolova, S. V., Morgunov, I. G., 2017. Metabolic peculiarities of the citric acid overproduction from glucose in yeasts Yarrowia lipolytica. Bioresource Technology. 243, 433440. Kamzolova, S. V., Yusupova, A. I., Vinokurova, N. G., Fedotcheva, N. I., Kondrashova, M. N., Finogenova, T. N., Morgunov, I. G., 2009. Chemically assisted microbial production of succinic acid by the yeast Yarrowia lipolytica grown on ethanol. Applied Microbiology and Biotechnology. 1027-1034. Katre, G., Ajmera, N., Zinjarde, S., RaviKumar, A., 2017. Mutants of Yarrowia lipolytica NCIM 3589 grown on waste cooking oil as a biofactory for biodiesel production. Microbial Cell Factories. 16, 176. Kavšček, M., Bhutada, G., Madl, T., Natter, K., 2015. Optimization of lipid production with a genome-scale model of Yarrowia lipolytica. BMC Systems Biology. 9, 72. Kawai, S., Hashimoto, W., Murata, K., 2010. Transformation of Saccharomyces cerevisiae and other fungi: methods and possible underlying mechanism. Bioengineered bugs. 1, 395-403. Kawasse, F. M., Amaral, P. F., Rocha-Leão, M. H. M., Amaral, A. L., Ferreira, E. C., Coelho, M. A. Z., 2003. Morphological analysis of Yarrowia lipolytica under stress conditions through image processing. Bioprocess and Biosystems Engineering. 25, 371-375. Kerkhoven, E. J., Pomraning, K. R., Baker, S. E., Nielsen, J., 2016. Regulation of aminoacid metabolism controls flux to lipid accumulation in Yarrowia lipolytica. NPJ systems biology and applications. 2, 16005. Kerkhoven, E. J., Kim, Y.-M., Wei, S., Nicora, C.D., Fillmore, T.L., Purvine, S.O., Webb-Robertson, B.J., Smith, R.D., Baker, S.E., Metz, T.O., Nielsen, J., 2017. Leucine Biosynthesis Is Involved in Regulating High Lipid Accumulation in Yarrowia lipolytica. mBio. 8. Kerscher, S., Durstewitz, G., Casaregola, S., Gaillardin, C., Brandt, U., 2001. The Complete mitochondrial genome of Yarrowia lipolytica. Comparative and Functional Genomics. 2. Kildegaard, K. R., Adiego-Pérez, B., Doménech Belda, D., Khangura, J. K., Holkenbrink, C., Borodina, I., 2017. Engineering of Yarrowia lipolytica for production of astaxanthin. Synthetic and Systems Biotechnology. 1-8. Knutsen, A. K., Robert, V., Poot, G. A., Epping, W., Figge, M., Holst-Jensen, A., Skaar, I., Smith, M. T., 2007. Polyphasic re-examination of Yarrowia lipolytica strains and the description of three novel Candida species: Candida oslonensis sp. nov., Candida alimentaria sp. nov. and Candida hollandica sp. nov. International Journal of Systematic and Evolutionary Microbiology. 57, 2426-2435. Kretzschmar, A., Otto, C., Holz, M., Werner, S., Hübner, L., Barth, G., 2013. Increased homologous integration frequency in Yarrowia lipolytica strains defective in non-homologous end-joining. Current Genetics. 59, 63-72. Kurtzman, C. P., 2011. Chapter 82 - Yarrowia van der Walt & von Arx (1980). The Yeasts (Fifth Edition). Elsevier, London, pp. 927-929.
53
Lane, S., Zhang, S., Wei, N., Rao, C., Jin, Y.S., 2015. Development and Physiological Characterization of Cellobiose-Consuming Yarrowia lipolytica. Biotechnology and Bioengineering. 112, 1012–22. Larroude, M., Celińska, E., Back, A., Thomas, S., Nicaud, J. M., Ledesma-Amaro, R., 2018. A synthetic biology approach to transform Yarrowia lipolytica into a competitive biotechnological producer of β-carotene. Biotechnology and Bioengineering. 115, 464-472. Lazar, Z., Dulermo, T., Neuvéglise, C., Coq, A.-m. C.-l., 2014. Hexokinase — A limiting factor in lipid production from fructose in Yarrowia lipolytica. Metabolic Engineering. 26, 8999. Lazar, Z., Rossignol, T., Verbeke, J., Crutz-Le Coq, A. M., Nicaud, J.-M., and Robak, M., 2013. Optimized invertase expression and secretion cassette for improving Yarrowia lipolytica growth on sucrose for industrial applications. Journal of Industrial Microbiology and Biotechnology. 40, 1273–83. Lazar, Z., Walczak, E., Robak, M., 2011. Simultaneous production of citric acid and invertase by Yarrowia lipolytica SUC+ transformants. Bioresource Technology. 102, 6982–89. Le Dall, M.-T., Nicaud, J.-M., Gaillardin, C., 1994. Multiple-copy integration in the yeast Yarrowia lipolytica. Current Genetics. 26, 38-44. Ledesma-Amaro, R., Nicaud, J.-M., 2016. Metabolic engineering for expanding the substrate range of Yarrowia lipolytica. Trends in Biotechnology. 34, 798–809. Le Hir, H., Nott, A., Moore, M. J., 2003. How introns influence and enhance eukaryotic gene expression. Trends in Biochemical Sciences. 28, 215-220. Leplat, C., Nicaud, J. M., Rossignol, T., 2015. High-throughput transformation method for Yarrowia lipolytica mutant library screening. FEMS Yeast Res. 15. Li, C., Yang, X., Gao, S., Chuh, A. H., Lin, C. S. K., 2018. Hydrolysis of fruit and vegetable waste for efficient succinic acid production with engineered Yarrowia lipolytica. Journal of Cleaner Production. 179, 151-159. Li, H., Alper, H. S., 2016. Enabling xylose utilization in Yarrowia lipolytica for lipid production. Biotechnology Journal. 11,1230–40. Li, Z. J., Qiao, K., Liu, N., Stephanopoulos, G., 2017. Engineering Yarrowia lipolytica for poly-3-hydroxybutyrate production. Journal of Industrial Microbiology and Biotechnology. 44, 605-612. Lian, J., Garcia-Perez, M., Coates, R., Wu, H., Chen, S., 2012. Yeast fermentation of carboxylic acids obtained from pyrolytic aqueous phases for lipid production. Bioresource Technology. 118, 177–86. Liu, H.-H., Madzak, C., Sun, M.-L., Ren, L.-J., Song, P., Huang, H., Ji, X.-J., 2017a. Engineering Yarrowia lipolytica for arachidonic acid production through rapid assembly of metabolic pathway. Biochemical Engineering Journal. 119, 52-58. Liu, H. H., Zeng, S. Y., Shi, T. Q., Ding, Y., Ren, L. J., Song, P., Huang, H., Madzak, C., Ji, X. J., 2017b. A Yarrowia lipolytica strain engineered for arachidonic acid production counteracts metabolic burden by redirecting carbon flux towards intracellular fatty acid accumulation at the expense of organic acids secretion. Biochemical Engineering Journal. 128, 201-209. Liu, L., Alper, H. S., 2014. Draft genome sequence of the oleaginous yeast Yarrowia lipolytica PO1f, a commonly used metabolic engineering host. Genome Announc. 2.
54
Liu, L., Markham, K., Blazeck, J., Zhou, N., Leon, D., Otoupal, P., Alper, H. S., 2015a. Surveying the lipogenesis landscape in Yarrowia lipolytica through understanding the function of a Mga2p regulatory protein mutant. Metabolic engineering. 31, 102-111. Liu, L., Otoupal, P., Pan, A., Alper, H. S., 2014. Increasing expression level and copy number of a Yarrowia lipolytica plasmid through regulated centromere function. FEMS Yeast Res. 14, 1124-7. Liu, L., Pan, A., Spofford, C., Zhou, N., Alper, H. S., 2015b. An evolutionary metabolic engineering approach for enhancing lipogenesis in Yarrowia lipolytica. Metabolic engineering. 29, 36-45. Liu, N., Qiao, K., Stephanopoulos, G., 2016. 13C Metabolic Flux Analysis of Acetate Conversion to Lipids by Yarrowia lipolytica. Metabolic Engineering 38, 86–97. Lustig, A. J., 1999. The Kudos of non-homologous end-joining. Nature Genetics. 23, 130. Ma, J.-L., Kim, E. M., Haber, J. E., Lee, S. E., 2003. Yeast Mre11 and Rad1 proteins define a Ku-independent mechanism to repair double-strand breaks lacking overlapping end sequences. Molecular and Cellular Biology. 23, 8820-8828. Madzak, C., Beckerich, J.-M., 2013. Heterologous protein expression and secretion in Yarrowia lipolytica. In: Barth, G., (Ed.), Yarrowia lipolytica: Biotechnological Applications. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 1-76. Madzak, C., Blanchin-Roland, S., Cordero Otero, R. R., Gaillardin, C., 1999. Functional analysis of upstream regulating regions from the Yarrowia lipolytica XPR2 promoter. Microbiology. 145, 75-87. Madzak, C., Gaillardin, C., Beckerich, J.-M., 2004. Heterologous protein expression and secretion in the non-conventional yeast Yarrowia lipolytica: a review. Journal of Biotechnology. 109, 63-81. Madzak, C., Treton, B., Blanchin-Roland, S., 2000. Strong hybrid promoters and integrative expression/secretion vectors for quasi-constitutive expression of heterologous proteins in the yeast Yarrowia lipolytica. J Mol Microbiol Biotechnol. 2, 207-16. Magnan, C., Yu, J., Chang, I., Jahn, E., Kanomata, Y., Wu, J., Zeller, M., Oakes, M., Baldi, P., Sandmeyer, S., 2016. Sequence assembly of Yarrowia lipolytica strain W29/CLIB89 shows transposable element diversity. PLOS One. 11, e0162363. Man, O., Pilpel, Y., 2007. Differential translation efficiency of orthologous genes is involved in phenotypic divergence of yeast species. Nature Genetics. 39, 415. Marchal, R., Vandecasteele, J.-P., Metche, M., 1977. Regulation of the central metabolism in relation to citric acid production in Saccharomycopsis lipolytica. Archives of Microbiology. 113, 99-104. Markham, K. A., Alper, H. S., 2018. Synthetic Biology Expands the Industrial Potential of Yarrowia lipolytica. Trends in Biotechnology. DOI: 10.1016/j.tibtech.2018.05.004 Markham, K. A., Cordova, L., Hill, A., Alper, H. S., 2017. Yarrowia lipolytica as a cell factory for oleochemical biotechnology. In: Lee, S. Y., (Ed.), Consequences of Microbial Interactions with Hydrocarbons, Oils, and Lipids: Production of Fuels and Chemicals. Springer International Publishing, Cham, pp. 1-18. Markham, K. A., Palmer, C. M., Chwatko, M., Wagner, J. M., Murray, C., Vazquez, S., Swaminathan, A., Chakravarty, I., Lynd, N. A., Alper, H. S., 2018. Rewiring Yarrowia lipolytica toward triacetic acid lactone for materials generation. Proceedings of the National Academy of Sciences. 115, 2096-2101. 55
Masoro, E. J. 1965. Mechanisms Related To the Homeostatic Regulation of Lipogenesis. Annals of the New York Academy of Sciences. 131,199–206. Matthäus, F., Ketelhot, M., Gatter, M., Barth, G., 2014. Production of lycopene in the non-carotenoid-producing yeast Yarrowia lipolytica. Applied and Environmental Microbiology. 80, 1660-1669. Mekouar, M., Blanc-Lenfle, I., Ozanne, C., Da Silva, C., Cruaud, C., Wincker, P., Gaillardin, C., Neuvéglise, C., 2010. Detection and analysis of alternative splicing in Yarrowia lipolytica reveal structural constraints facilitating nonsense-mediated decay of intron-retaining transcripts. Genome Biology. 11, R65. Michely, S., Gaillardin, C., Nicaud, J.-M., Neuvéglise, C., 2013. Comparative physiology of oleaginous species from the Yarrowia clade. PLOS One. 8, e63356. Mirończuk, A.M., Rzechonek, D.A., Biegalska, A., Rakicka, M., Dobrowolski, A., 2016. A Novel Strain of Yarrowia lipolytica as a Platform for Value-Added Product Synthesis from Glycerol. Biotechnology for Biofuels 9, 1–12. Mirończuk, A. M., Biegalska, A., Dobrowolski, A., 2017. Functional overexpression of genes involved in erythritol synthesis in the yeast Yarrowia lipolytica. Biotechnology for Biofuels. 10, 1-12. Mishra, P., Lee, N. R., Lakshmanan, M., Kim, M., Kim, B. G., Lee, D. Y., 2018. Genome-scale model-driven strain design for dicarboxylic acid production in Yarrowia lipolytica. BMC Syst Biol. 12, 12. Moeller, L., Zehnsdorf, A., Aurich, A., Barth, G., Bley, T., Strehlitz, B., 2013. Citric Acid Production from Sucrose by Recombinant Yarrowia lipolytica Using Semicontinuous Fermentation. Engineering in Life Sciences. 13,163–71. Morales-Vargas, A. T., Domínguez, A., Ruiz-Herrera, J., 2012. Identification of dimorphism-involved genes of Yarrowia lipolytica by means of microarray analysis. Research in Microbiology. 163, 378-387. Morgunov, I. G., Kamzolova, S. V., Perevoznikova, O. A., Shishkanova, N. V., Finogenova, T. V., 2004. Pyruvic acid production by a thiamine auxotroph of Yarrowia lipolytica. Process Biochemistry. 39, 1469-1474. Mori, K., Iwama, R., Kobayashi, S., Horiuchi, H., Fukuda, R., Ohta, A., 2013. Transcriptional repression by glycerol of genes involved in the assimilation of N-alkanes and fatty acids in yeast Yarrowia lipolytica. FEMS Yeast Research. 13, 233–40. Morin, N., Cescut, J., Beopoulos, A., Lelandais, G., Le Berre, V., Uribelarrea, J.-L., Molina-Jouve, C., Nicaud, J.-M., 2011. Transcriptomic analyses during the transition from biomass production to lipid accumulation in the oleaginous yeast Yarrowia lipolytica. PLOS One. 6, e27966. Morse, N. J., Wagner, J. M., Reed, K. B., Gopal, M. R., Lauffer, L. H., Alper, H. S., 2018. T7 Polymerase expression of guide RNAs in vivo allows exportable CRISPR-Cas9 editing in multiple yeast hosts. ACS Synth Biol. 7, 1075-1084. Nambou, K., Jian, X., Zhang, X., Wei, L., Lou, J., Madzak, C., Hua, Q., 2015. Flux balance analysis inspired bioprocess upgrading for lycopene production by a metabolically engineered strain of Yarrowia lipolytica. 794-813. Neuvéglise, C., Chalvet, F., Wincker, P., Gaillardin, C., Casaregola, S., 2005. Mutatorlike element in the yeast Yarrowia lipolytica displays multiple alternative splicings. Eukaryotic Cell. 4, 615-624.
56
Neuvéglise, C., Gaillardin, C., Marck, C., 2013. Noncoding RNA genes transcribed by RNA polymerase III in Yarrowia lipolytica. In: Barth, G., (Ed.), Yarrowia lipolytica: Genetics, Genomics, and Physiology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 79-109. Nicaud, J.-M., Gaillardin, C., Pignede, G., Method For non-homologous transformation of Yarrowia lipolytica. Agronomique Inst Nat Rech Centre Nat Rech Scient, US, 2003. Nicaud, J.-M., Fabre, E., Gaillardin, C., 1989. Expression of invertase activity in Yarrowia lipolytica and its use as a selective marker. Curr Genet. 16, 253-60. Nicaud, J.-M., Madzak, C., van den Broek, P., Gysler, C., Duboc, P., Niederberger, P., Gaillardin, C., 2002. Protein expression and secretion in the yeast Yarrowia lipolytica. FEMS Yeast Res. 2, 371-9. Niehus, X., Crutz-Le Coq, A.-M., Sandoval, G., Nicaud, J.-M., Ledesma-Amaro, R., 2018. Engineering Yarrowia lipolytica to enhance lipid production from lignocellulosic materials. Biotechnology for Biofuels. 11, 11. O'Shea, D. G., Walsh, P. K., 2000. The effect of culture conditions on the morphology of the dimorphic yeast Kluyveromyces marxianus var. marxianus NRRLy2415: a study incorporating image analysis. Applied Microbiology and Biotechnology. 53, 316-322. Pagot, Y., Endrizzi, A., Nicaud, J., Belin, J., 1997. Utilization of an auxotrophic strain of the yeast Yarrowia lipolytica to improve gamma-decalactone production yields. Letters in Applied Microbiology. 25, 113-116. Paiva, S., Devaux, F., Barbosa, S., Jacq, C., Casal, M., 2004. Ady2p is essential for the acetate permease activity in the yeast Saccharomyces cerevisiae. Yeast. 21, 201-210. Papanikolaou, S., Muniglia, L., Chevalot, I., Aggelis G., Marc, I., 2002. Yarrowia lipolytica as a potential producer of citric acid from raw glycerol. Journal of Applied Microbiology. 92, 737–44. Papanikolaou, S., Aggelis, G., 2002. Lipid production by Yarrowia lipolytica growing on industrial glycerol in a single stage continuous culture. Bioresource Technology. 82, 43–49. Patterson, K., Yu, J., Landberg, J., Chang, I., Shavarebi, F., Bilanchone, V., Sandmeyer, S., 2018. Functional genomics for the oleaginous yeast Yarrowia lipolytica. Metabolic engineering. Pérez-Campo, F. M., Domínguez, A., 2001. Factors affecting the morphogenetic switch in Yarrowia lipolytica. Current Microbiology. 43, 429-433. Petit, T., Gancedo, C., 1999. Molecular cloning and characterization of the gene HXK1 encoding the hexokinase from Yarrowia lipolytica. Yeast. 15, 1573–84. Pfleger, B. F., 2016. Microbes paired for biological gas-to-liquids (Bio-GTL) process. Proceedings of the National Academy of Sciences. 113, 3717–19. Pfleger, B. F., Gossing, M., Nielsen, J., 2015. Metabolic engineering strategies for microbial synthesis of oleochemicals. Metabolic Engineering. 29, 1–11. Pignède, G., Wang, H.-J., Fudalej, F., Seman, M., Gaillardin, C., Nicaud, J.-M., 2000. Autocloning and amplification of LIP2 in Yarrowia lipolytica. Applied and Environmental Microbiology. 66, 3283-3289. Pomraning, K. R., Baker, S. E., 2015. Draft genome sequence of the dimorphic yeast Yarrowia lipolytica strain W29. Genome Announcements. 3. Qiao, K., Imam Abidi, S. H., Liu, H., Zhang, H., Chakraborty, S., Watson, N., Kumaran Ajikumar, P., Stephanopoulos, G., 2015. Engineering lipid overproduction in the oleaginous yeast Yarrowia lipolytica. Metabolic Engineering. 29, 56-65.
57
Qiao, K., Wasylenko, T. M., Zhou, K., Xu, P., Stephanopoulos, G., 2017. Lipid production in Yarrowia lipolytica is maximized by engineering cytosolic redox metabolism. Nat Biotech. 35, 173-177. Quarterman, J., Slininger, P. J., Kurtzman, C. P., Thompson, S. R., Dien, B. S., 2017. A survey of yeast from the Yarrowia clade for lipid production in dilute acid pretreated lignocellulosic biomass hydrolysate. Applied Microbiology and Biotechnology. 101, 3319-3334. Rakicka, M., Lazar, Z., Dulermo, T., Fickers, P., Nicaud, J.-M., 2015. Lipid production by the oleaginous yeast Yarrowia lipolytica using industrial by-products under different culture conditions. Biotechnology for Biofuels. 8, 104. Rakicka, M., Biegalska, A., Rymowicz, W., Dobrowolski, A., Mirończuk, A. M., 2017. Polyol production from waste materials by genetically modified Yarrowia lipolytica. Bioresource Technology. 243, 393-399. Ratledge, C., Wynn, J.P., 2002. The biochemistry and molecular biology of lipid accumulation in oleaginous microorganisms. Advances in Applied Microbiology. 51, 1–51. Richard, G.-F., Kerrest, A., Lafontaine, I., Dujon, B., 2005. Comparative genomics of hemiascomycete yeasts: Genes involved in DNA replication, repair, and recombination. Molecular Biology and Evolution. 22, 1011-1023. Robles-Rodriguez, C. E., Munoz-Tamayo, R., Bideaux, C., Gorret, N., Guillouet, S. E., Molina-Jouve, C., Roux, G., Aceves-Lara, C. A., 2018. Modeling and optimization of lipid accumulation by Yarrowia lipolytica from glucose under nitrogen depletion conditions. Biotechnol Bioeng. 115, 1137-1151. Rodriguez, G. M., Shabbir-Hussain, M., Gambill, L., Gao, D., Yaguchi, A., Blenner, M., 2016. Engineering xylose utilization in Yarrowia lipolytica by understanding its cryptic xylose pathway. Biotechnology for Biofuels. 9, 149. Roostita, R., Fleet, G. H., 1996. The occurrence and growth of yeasts in camembert and blue-veined cheeses. International Journal of Food Microbiology. 28, 393-404. Ruiz-Herrera, J., Sentandreu, R., 2002. Different effectors of dimorphism in Yarrowia lipolytica. Archives of Microbiology. 178, 477-483. Rymowicz, W., Rywińska, A., Gładkowski, W., 2008. Simultaneous production of citric acid and erythritol from crude glycerol by Yarrowia lipolytica Wratislavia K1. Chemical Papers. 62, 239–46. Rymowicz, W., Rywińska, A., Marcinkiewicz, M., 2009. High-yield production of erythritol from raw glycerol in fed-batch cultures of Yarrowia lipolytica. Biotechnology Letters. 31, 377-380. Sabra, W., Bommareddy, R. R., Maheshwari, G., Papanikolaou, S., Zeng, A. P., 2017. Substrates and oxygen dependent citric acid production by Yarrowia lipolytica: Insights through transcriptome and fluxome analyses. Microbial Cell Factories. 16, 1-14. Sagnak, R., Cochot, S., Molina-Jouve, C., Nicaud, J. M., Guillouet, S. E., 2018. Modulation of the glycerol phosphate availability led to concomitant reduction in the citric acid excretion and increase in lipid content and yield in Yarrowia lipolytica. Journal of Biotechnology. 265, 40-45. Savická, D., Šilhánková, L., 1995. Drug resistance in Yarrowia lipolytica. Folia Microbiologica. 40, 89-94. Schmid-Berger, N., Schmid, B., Barth, G., 1994. Ylt1, a highly repetitive retrotransposon in the genome of the dimorphic fungus Yarrowia lipolytica. J Bacteriol. 176, 2477-82.
58
Schwartz, C., Curtis, N., Lobs, A. K., Wheeldon, I., 2018. Multiplexed CRISPR Activation of cryptic sugar metabolism enables Yarrowia lipolytica growth on cellobiose. Biotechnol J. e1700584. Schwartz, C., Frogue, K., Misa, J., Wheeldon, I., 2017a. Host and pathway engineering for enhanced lycopene biosynthesis in Yarrowia lipolytica. Frontiers in Microbiology. 8, 1-11. Schwartz, C., Shabbir-Hussain, M., Frogue, K., Blenner, M., Wheeldon, I., 2017b. Standardized markerless gene integration for pathway engineering in Yarrowia lipolytica. ACS Synthetic Biology. 6, 402-409. Schwartz, C., Wheeldon, I., 2018. CRISPR-Cas9-mediated genome editing and transcriptional control in Yarrowia lipolytica. Methods in molecular biology (Clifton, N.J.). 1772, 327-345. Schwartz, C. M., Shabbir-Hussain, M., Blenner, M., Wheeldon, I., 2016. Synthetic RNA polymerase III promoters facilitate high-efficiency CRISPR-Cas9-mediated genome editing in Yarrowia lipolytica. ACS Synth Biol. 5, 356-9. Shabbir Hussain, M., Gambill, L., Smith, S., Blenner, M. A., 2016. Engineering promoter architecture in oleaginous yeast Yarrowia lipolytica. ACS Synthetic Biology. 5, 213-223. Shabbir Hussain, M., Wheeldon, I., Blenner, M. A., 2017. A strong hybrid fatty acid inducible transcriptional sensor built from Yarrowia lipolytica upstream activating and regulatory sequences. Biotechnology Journal. 12, 1-11. Silverman, A. M., Qiao, K., Xu, P., Stephanopoulos, G., 2016. Functional overexpression and characterization of lipogenesis-related genes in the oleaginous yeast Yarrowia lipolytica. Applied Microbiology and Biotechnology. 100, 3781–98. Stajich, J. E., Dietrich, F. S., Roy, S. W., 2007. Comparative genomic analysis of fungal genomes reveals intron-rich ancestors. Genome Biology. 8, R223. Sumita, T., Iida, T., Yamagami, S., Horiuchi, H., Takagi, M., Ohta, A., 2002. YlALK1 encoding the cytochrome P450ALK1 in Yarrowia lipolytica is transcriptionally induced by nalkane through two distinct cis-elements on its promoter. Biochemical and Biophysical Research Communications. 294, 1071-1078. Sun, M. L., Madzak, C., Liu, H. H., Song, P., Ren, L. J., Huang, H., Ji, X. J., 2017. Engineering Yarrowia lipolytica for efficient γ-linolenic acid production. Biochemical Engineering Journal. 117, 172-180. Tai, M., Stephanopoulos, G., 2013. Engineering the push and pull of lipid biosynthesis in oleaginous yeast Yarrowia lipolytica for biofuel production. Metabolic Engineering. 15, 1-9. Tehlivets, O., Scheuringer, K., Kohlwein, S. D., 2007. Fatty acid synthesis and elongation in yeast. Biochimica et Biophysica Acta - Molecular and Cell Biology of Lipids. 1771, 255–70. Tenagy, Park, J.S., Iwama, R., Kobayashi, S., Ohta, A., Horiuchi, H., Fukuda, R., 2015. Involvement of acyl-CoA synthetase genes in N-alkane assimilation and fatty acid utilization in yeast Yarrowia lipolytica. FEMS Yeast Research. 15, 1–12. Thevenieau, F., Beopoulos, A., Desfougeres, T., Sabirova, J., Albertin, K., Zinjarde, S., Nicaud, J.-M., 2010. Uptake and assimilation of hydrophobic substrates by the oleaginous yeast Yarrowia lipolytica. In: Timmis, K. N., (Ed.), Handbook of Hydrocarbon and Lipid Microbiology. Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 1513-1527. Thevenieau, F., Nicaud, J.-M., Gaillardin, C., 2009. Applications of the non-conventional yeast Yarrowia lipolytica. In: Satyanarayana, T., Kunze, G., Eds.), Yeast Biotechnology: Diversity and Applications. Springer Netherlands, Dordrecht, pp. 589-613. 59
Timoumi, A., Cléret, M., Bideaux, C., Guillouet, S. E., Allouche, Y., Molina-Jouve, C., Fillaudeau, L., Gorret, N., 2017. Dynamic behavior of Yarrowia lipolytica in response to pH perturbations: dependence of the stress response on the culture mode. Applied Microbiology and Biotechnology. 101, 351-366. Torres-Guzmán, J. C., Domínguez, A., 1997. HOY1, a homeo gene required for hyphal formation in Yarrowia lipolytica. Molecular and Cellular Biology. 17, 6283-93. Trassaert, M., Vandermies, M., Carly, F., Denies, O., Thomas, S., Fickers, P., Nicaud, J.M., 2017. New inducible promoter for gene expression and synthetic biology in Yarrowia lipolytica. Microbial Cell Factories. 16, 141. Trebulle, P., Nicaud, J. M., Leplat, C., Elati, M., 2017. Inference and interrogation of a coregulatory network in the context of lipid accumulation in Yarrowia lipolytica. NPJ systems biology and applications. 3, 21. Verbeke, J., Beopoulos, A., Nicaud, J.-M., 2013. Efficient homologous recombination with short length flanking fragments in Ku70 deficient Yarrowia lipolytica strains. Biotechnology Letters. 35, 571-576. Vernis, L., Abbas, A., Chasles, M., Gaillardin, C. M., Brun, C., Huberman, J. A., Fournier, P., 1997. An origin of replication and a centromere are both needed to establish a replicative plasmid in the yeast Yarrowia lipolytica. Mol Cell Biol. 17, 1995-2004. Vernis, L., Poljak, L., Chasles, M., Uchida, K., Casaregola, S., Kas, E., Matsuoka, M., Gaillardin, C., Fournier, P., 2001. Only centromeres can supply the partition system required for ARS function in the yeast Yarrowia lipolytica. J Mol Biol. 305, 203-17. Wache, Y., Pagot, Y., Nicaud, J.-M., Belin, J.-M., 1998. Acyl-CoA oxidase , a key step for lactone production by Yarrowia lipolytica. Journal of Molecular Catalysis B: Enzymatic. 5, 165-169. Wagner, J. M., Liu, L., Yuan, S.-f., Venkataraman, M. V., Abate, A. R., Alper, H. S., 2018a. A comparative analysis of single cell and droplet-based FACS for improving production phenotypes : Riboflavin overproduction in Yarrowia lipolytica. Metabolic Engineering. 47, 346356. Wagner, J. M., Williams, E. V., Alper, H. S., 2018b. Developing a piggyBac transposon system and compatible selection markers for insertional mutagenesis and genome engineering in Yarrowia lipolytica. Biotechnol J. 13, e1800022. Wang, G., Li, D., Miao, Z., Zhang, S., Liang, W., Liu, L., 2017. Comparative transcriptome analysis reveals multiple functions for Mhy1p in lipid biosynthesis in the oleaginous yeast Yarrowia lipolytica. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1863, 81-90. Wang, G., Xiong, X., Ghogare, R., Wang, P., Meng, Y., Chen, S., 2016. Exploring fatty alcohol-producing capability of Yarrowia lipolytica. Biotechnology for Biofuels. 9, 107. Wang, H. J., Le Dall, M.T., Wache, Y., Laroche, C., Belin, J.M., Gaillardin, C., Nicaud, J.-M., 1999. Evaluation of acyl coenzyme A oxidase (Aox) isozyme function in the N-alkaneassimilating yeast Yarrowia lipolytica. Journal of Bacteriology. 181, 5140–48. Wang, J. H., Hung, W., Tsai, S. H., 2011. High efficiency transformation by electroporation of Yarrowia lipolytica. Journal of microbiology (Seoul, Korea). 49, 469-72. Wasylenko, T. M., Ahn, W. S., Stephanopoulos, G., 2015. The oxidative pentose phosphate pathway is the primary source of NADPH for lipid overproduction from glucose in Yarrowia lipolytica. Metabolic Engineering. 30, 27-39.
60
Wei, S., Jian, X., Chen, J., Zhang, C., Hua, Q., 2017. Reconstruction of genome-scale metabolic model of Yarrowia lipolytica and its application in overproduction of triacylglycerol. Bioresources and Bioprocessing. 4, 51. Wong, L., Engel, J., Jin, E., Holdridge, B., Xu, P., 2017. YaliBricks , a versatile genetic toolkit for streamlined and rapid pathway engineering in Yarrowia lipolytica. Metabolic Engineering Communications. 5, 68-77. Wynn, J. P., Kendrick, A., Ratledge, C., 1997. Sesamol as an inhibitor of growth and lipid metabolism in Mucor circinelloides via its action on malic enzyme. Lipids. 32, 605–10. Xie, D., Jackson, E. N., Zhu, Q., 2015. Sustainable source of omega-3 eicosapentaenoic acid from metabolically engineered Yarrowia lipolytica: from fundamental research to commercial production. Applied Microbiology and Biotechnology. 99, 1599-1610. Xie, D., Miller, E., Sharpe, P., Jackson, E., Zhu, Q., 2017. Omega-3 production by fermentation of Yarrowia lipolytica: From fed-batch to continuous. Biotechnology and Bioengineering. 114, 798-812. Xu, P., Qiao, K., Ahn, W. S., Stephanopoulos, G., 2016. Engineering Yarrowia lipolytica as a platform for synthesis of drop-in transportation fuels and oleochemicals. Proceedings of the National Academy of Sciences. 113, 10848-10853. Xu, P., Qiao, K., Stephanopoulos, G., 2017. Engineering oxidative stress defense pathways to build a robust lipid production platform in Yarrowia lipolytica. Biotechnology and Bioengineering. 114, 1521-1530. Xue, Z., Sharpe, P. L., Hong, S.-P., Yadav, N. S., Xie, D., Short, D. R., Damude, H. G., Rupert, R. a., Seip, J. E., Wang, J., Pollak, D. W., Bostick, M. W., Bosak, M. D., Macool, D. J., Hollerbach, D. H., Zhang, H., Arcilla, D. M., Bledsoe, S. a., Croker, K., McCord, E. F., Tyreus, B. D., Jackson, E. N., Zhu, Q., 2013. Production of omega-3 eicosapentaenoic acid by metabolic engineering of Yarrowia lipolytica. Nature biotechnology. 31, 734-40. Yamane, T., Ogawa, T., Matsuoka, M., 2008a. Derivation of consensus sequence for protein binding site in Yarrowia lipolytica centromere. Journal of Bioscience and Bioengineering. 105, 671-674. Yamane, T., Sakai, H., Nagahama, K., Ogawa, T., Matsuoka, M., 2008b. Dissection of centromeric DNA from yeast Yarrowia lipolytica and identification of protein-binding site required for plasmid transmission. Journal of Bioscience and Bioengineering. 105, 571-578. Yang, F., Hanna, M. A., Sun, R., 2012. Value-added uses for crude glycerol--a byproduct of biodiesel production. Biotechnology for Biofuels. 5, 13. Yang, X., Nambou, K., Wei, L., Hua, Q., 2016. Heterologous production of α-farnesene in metabolically engineered strains of Yarrowia lipolytica. Bioresource Technology. 216, 10401048. Yang, X., Wang, H., Li, C., Lin, C. S. K., 2017. Restoring of glucose metabolism of engineered Yarrowia lipolytica for succinic acid production via a simple and efficient adaptive evolution strategy. Journal of Agricultural and Food Chemistry. 65, 4133-4139. Yovkova, V., Otto, C., Aurich, A., Mauersberger, S., Barth, G., 2014. Engineering the alpha-ketoglutarate overproduction from raw glycerol by overexpression of the genes encoding NADP+-dependent isocitrate dehydrogenase and pyruvate carboxylase in Yarrowia lipolytica. Applied Microbiology and Biotechnology. 98, 2003-2013. Yu, J., Landberg, J., Shavarebi, F., Bilanchone, V., Okerlund, A., Wanninayake, U., Zhao, L., Kraus, G., Sandmeyer, S., 2018. Bioengineering triacetic acid lactone production in Yarrowia lipolytica for pogostone synthesis. Biotechnology and Bioengineering. 61
Yuzbasheva, E. Y., Mostova, E. B., Andreeva, N. I., Yuzbashev, T. V., Fedorov, A. S., Konova, I. A., Sineoky, S. P., 2017a. A metabolic engineering strategy for producing free fatty acids by the Yarrowia lipolytica yeast based on impairment of glycerol metabolism. Biotechnology and Bioengineering. 1-11. Yuzbasheva, E. Y., Mostova, E. B., Andreeva, N. I., Yuzbashev, T. V., Laptev, I. A., Sobolevskaya, T. I., Sineoky, S. P., 2017b. Co-expression of glucose-6-phosphate dehydrogenase and acyl-CoA binding protein enhances lipid accumulation in the yeast Yarrowia lipolytica. New Biotechnology. 39, 18-21. Zeng, W., Du, G., Chen, J., Li, J., Zhou, J., 2015. A high-throughput screening procedure for enhancing alpha-ketoglutaric acid production in Yarrowia lipolytica by random mutagenesis. Process Biochemistry. 50, 1516-1522. Zeng, W., Fang, F., Liu, S., Du, G., Chen, J., Zhou, J., 2016. Comparative genomics analysis of a series of Yarrowia lipolytica WSH-Z06 mutants with varied capacity for alphaketoglutarate production. Journal of Biotechnology. 239, 76-82. Zeng, W., Zhang, H., Xu, S., Fang, F., Zhou, J., 2017. Biosynthesis of keto acids by fedbatch culture of Yarrowia lipolytica WSH-Z06. Bioresource Technology. 243, 1037-1043. Zhang, H., Zhang, L., Chen, H., Chen, Y.Q., Ratledge, C., Song, Y., Chen, W., 2013. Regulatory properties of malic enzyme in the oleaginous yeast, Yarrowia lipolytica, and its noninvolvement in lipid accumulation. Biotechnology Letters. 35, 2091–98. Zhang, J. L., Peng, Y. Z., Liu, D., Liu, H., Cao, Y. X., Li, B. Z., Li, C., Yuan, Y. J., 2018. Gene repression via multiplex gRNA strategy in Y. lipolytica. Microb Cell Fact. 17, 62. Zhang, Y., Adams, I.P., Ratledge, C., 2007. Malic enzyme: the controlling activity for lipid production? Overexpression of malic enzyme in Mucor circinelloides leads to a 2.5-fold increase in lipid accumulation. Microbiology, 153, 2013–25. Zhang, Y., Wang, Y., Yao, M., Liu, H., Zhou, X., Xiao, W., Yuan, Y., 2017. Improved campesterol production in engineered Yarrowia lipolytica strains. Biotechnology Letters. Zhao, C., Gao, Q., Chen, J., Wei, L., Imanaka, T., Hua, Q., 2017. Metabolomic changes and metabolic responses to expression of heterologous biosynthetic genes for lycopene production in Yarrowia lipolytica. J Biotechnol. 251, 174-185. Zhou, J., Yin, X., Madzak, C., Du, G., Chen, J., 2012. Enhanced alpha-ketoglutarate production in Yarrowia lipolytica WSH-Z06 by alteration of the acetyl-CoA metabolism. Journal of Biotechnology. 161, 257-264.
62
63
64
Highlights:
Yarrowia lipolytica is a host for both lipid and nonlipid chemical products Emerging synthetic biology tools enable further engineering in this host Successful engineering for lipid production phenotypes Engineering for both native and nonnative lipid, organic acid, sugar and acetyl-CoA derived products
65